25.09.2013 Views

Color Atlas of Hematology (2004).pdf

Color Atlas of Hematology (2004).pdf

Color Atlas of Hematology (2004).pdf

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

i


ii<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


<strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong><br />

Practical Microscopic and Clinical Diagnosis<br />

Harald Theml, M.D.<br />

Pr<strong>of</strong>essor, Private Practice<br />

<strong>Hematology</strong>/Oncology<br />

Munich, Germany<br />

Heinz Diem, M.D.<br />

Klinikum Grosshadern<br />

Institute <strong>of</strong> Clinical Chemistry<br />

Munich, Germany<br />

Torsten Haferlach, M.D.<br />

Pr<strong>of</strong>essor, Klinikum Grosshadern<br />

Laboratory for Leukemia Diagnostics<br />

Munich, Germany<br />

2nd revised edition<br />

262 color illustrations<br />

32 tables<br />

Thieme<br />

Stuttgart · New York<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

iii


iv<br />

Library <strong>of</strong> Congress Cataloging-in-Publication<br />

Data is available from the publisher<br />

This book is an authorized revised<br />

translation <strong>of</strong> the 5th German edition<br />

published and copyrighted 2002 by<br />

Thieme Verlag, Stuttgart, Germany.<br />

Title <strong>of</strong> the German edition:<br />

Taschenatlas der Hämatologie<br />

Translator: Ursula Peter-Czichi PhD,<br />

Atlanta, GA, USA<br />

1st German edition 1983<br />

2nd German edition 1986<br />

3rd German edition 1991<br />

4th German edition 1998<br />

5th German edition 2002<br />

1st English edition 1985<br />

1st French edition 1985<br />

2nd French edition 2000<br />

1st Indonesion edition 1989<br />

1st Italian edition 1984<br />

1st Japanese edition 1997<br />

<strong>2004</strong> Georg Thieme Verlag<br />

Rüdigerstraße 14, 70469 Stuttgart,<br />

Germany<br />

http://www.thieme.de<br />

Thieme New York, 333 Seventh Avenue,<br />

New York, NY 10001 USA<br />

http://www.thieme.com<br />

Cover design: Cyclus, Stuttgart<br />

Typesetting and printing in Germany by<br />

Druckhaus Götz GmbH, Ludwigsburg<br />

ISBN 3-13-673102-6 (GTV)<br />

ISBN 1-58890-193-9 (TNY) 1 2 3 4 5<br />

Important note: Medicine is an everchanging<br />

science undergoing continual<br />

development. Research and clinical experience<br />

are continually expanding our<br />

knowledge, in particular our knowledge <strong>of</strong><br />

proper treatment and drug therapy. Ins<strong>of</strong>ar<br />

as this book mentions any dosage or application,<br />

readers may rest assured that the<br />

authors, editors, and publishers have made<br />

every effort to ensure that such references<br />

are in accordance with the state <strong>of</strong> knowledge<br />

at the time <strong>of</strong> production <strong>of</strong> the<br />

book.<br />

Nevertheless, this does not involve, imply,<br />

or express any guarantee or responsibility<br />

on the part <strong>of</strong> the publishers in respect to<br />

any dosage instructions and forms <strong>of</strong> applications<br />

stated in the book. Every user is requested<br />

to examine carefully the manufacturers’<br />

leaflets accompanying each drug<br />

and to check, if necessary in consultation<br />

with a physician or specialist, whether the<br />

dosage schedules mentioned therein or the<br />

contraindications stated by the manufacturers<br />

differ from the statements made in<br />

the present book. Such examination is particularly<br />

important with drugs that are<br />

either rarely used or have been newly released<br />

on the market. Every dosage<br />

schedule or every form <strong>of</strong> application used<br />

is entirely at the user’s own risk and responsibility.<br />

The authors and publishers request<br />

every user to report to the publishers<br />

any discrepancies or inaccuracies noticed.<br />

Some <strong>of</strong> the product names, patents, and<br />

registered designs referred to in this book<br />

are in fact registered trademarks or proprietary<br />

names even though specific reference<br />

to this fact is not always made in the<br />

text. Therefore, the appearance <strong>of</strong> a name<br />

without designation as proprietary is not to<br />

be construed as a representation by the<br />

publisher that it is in the public domain.<br />

This book, including all parts there<strong>of</strong>, is legally<br />

protected by copyright. Any use, exploitation,<br />

or commercialization outside<br />

the narrow limits set by copyright legislation,<br />

without the publisher’s consent, is illegal<br />

and liable to prosecution. This applies in<br />

particular to photostat reproduction, copying,<br />

mimeographing, preparation <strong>of</strong> micr<strong>of</strong>ilms,<br />

and electronic data processing and<br />

storage.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Preface<br />

Our Current Edition<br />

Although this is the second English edition <strong>of</strong> our hematology atlas, this<br />

edition is completely new. As an immediate sign <strong>of</strong> this change, there are<br />

now three authors. The completely updated visual presentation uses digital<br />

images, and the content is organized according to the most up-to-date<br />

morphological classification criteria.<br />

In this new edition, our newly formed team <strong>of</strong> authors from Munich<br />

(the “Munich Group”) has successfully shared their knowledge with you.<br />

Heinz Diem and Torsten Haferlach are nationally recognized as lecturers<br />

<strong>of</strong> the diagnostics curriculum <strong>of</strong> the German Association for <strong>Hematology</strong><br />

and Oncology.<br />

Goals<br />

Most physicians are fundamentally “visually oriented.” Apart from immediate<br />

patient care, the microscopic analysis <strong>of</strong> blood plays to this preference.<br />

This explains the delight and level <strong>of</strong> involvement on the part <strong>of</strong><br />

practitioners in the pursuit <strong>of</strong> morphological analyses.<br />

Specialization notwithstanding, the hematologist wants to preserve<br />

the opportunity to perform groundbreaking diagnostics in hematology for<br />

the general practitioner, surgeon, pediatrician, the MTA technician, and all<br />

medical support personnel. New colleagues must also be won to the<br />

cause. Utmost attention to the analysis <strong>of</strong> hematological changes is essential<br />

for a timely diagnosis.<br />

Even before bone marrow cytology, cytochemistry, or immunocytochemistry,<br />

information based on the analysis <strong>of</strong> blood is <strong>of</strong> immediate relevance<br />

in the doctor’s <strong>of</strong>fice. It is central to the diagnosis <strong>of</strong> the diseases <strong>of</strong><br />

the blood cell systems themselves, which make their presence known<br />

through changes in blood components.<br />

The exhaustive quantitative and qualitative use <strong>of</strong> hematological diagnostics<br />

is crucial. Discussions with colleagues from all specialties and<br />

teaching experience with advanced medical students confirm its importance.<br />

In cases where a diagnosis remains elusive, the awareness <strong>of</strong> the<br />

next diagnostic step becomes relevant. Then, further investigation<br />

through bone marrow, lymph node, or organ tissue cytology can yield firm<br />

results. This pocket atlas <strong>of</strong>fers the basic knowledge for the use <strong>of</strong> these<br />

techniques as well.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

v


vi<br />

Preface<br />

Organization<br />

Reflecting our goals, the inductive organization proceeds from simple to<br />

specialized diagnostics. By design, we subordinated the description <strong>of</strong> the<br />

bone marrow cytology to the diagnostic blood analysis (CBC). However,<br />

we have responded to feedback from readers <strong>of</strong> the previous editions and<br />

have included the principles <strong>of</strong> bone marrow diagnostics and nonambiguous<br />

clinical bone marrow findings so that frequent and relevant<br />

diagnoses can be quickly made, understood, or replicated.<br />

The nosology and differential diagnosis <strong>of</strong> hematological diseases are<br />

presented to you in a tabular form. We wanted to <strong>of</strong>fer you a pocketbook<br />

for everyday work, not a reference book. Therefore, morphological curiosities,<br />

or anomalies, are absent in favor <strong>of</strong> a practical approach to morphology.<br />

The cellular components <strong>of</strong> organ biopsies and exudates are<br />

briefly discussed, mostly as a reminder <strong>of</strong> the importance <strong>of</strong> these tests.<br />

The images are consistently photographed as they normally appear in<br />

microscopy (magnification 100 or 63 with oil immersion lens, occasionally<br />

master-detail magnification objective 10 or 20). Even though<br />

surprising perspectives sometimes result from viewing cells at a higher<br />

magnification, the downside is that this by no means facilitates the recognition<br />

<strong>of</strong> cells using your own microscope.<br />

Instructions for the Use <strong>of</strong> this <strong>Atlas</strong><br />

The organization <strong>of</strong> this atlas supports a systematic approach to the study<br />

<strong>of</strong> hematology (see Table <strong>of</strong> Contents). The index <strong>of</strong>fers ways to answer<br />

detailed questions and access the hematological terminology with references<br />

to the main description and further citations.<br />

The best way to become familiar with your pocket atlas is to first have a<br />

cursory look through its entire content. The images are accompanied by<br />

short legends. On the pages opposite the images you will find corresponding<br />

short descriptive texts and tables. This text portion describes cell phenomena<br />

and discusses in more detail further diagnostic steps as well as<br />

the diagnostic approach to disease manifestations.<br />

Acknowledgments<br />

Twenty years ago, Pr<strong>of</strong>essor Herbert Begemann dedicated the foreword to<br />

the first edition <strong>of</strong> this hematology atlas. He acknowledged that—beyond<br />

cell morphology—this atlas aims at the clinical picture <strong>of</strong> patients. We are<br />

grateful for being able to continue this tradition, and for the impulses from<br />

our teachers and companions that make this possible.<br />

We thank our colleagues: J. Rastetter, W. Kaboth, K. Lennert, H. Löffler,<br />

H. Heimpel, P.M. Reisert, H. Brücher, W. Enne, T. Binder, H.D. Schick, W.<br />

Hiddemann, D. Seidel.<br />

Munich, January <strong>2004</strong> Harald Theml, Heinz Diem, Torsten Haferlach<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Contents<br />

vii<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells:<br />

Methods and Test Procedures . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1<br />

Introduction to the Physiology and Pathophysiology <strong>of</strong> the<br />

Hematopoietic System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2<br />

Cell Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2<br />

Principles <strong>of</strong> Regulation and Dysregulation in the Blood Cell<br />

Series and their Diagnostic Implications . . . . . . . . . . . . . . . . . . . . . . . . 7<br />

Procedures, Assays, and Normal Values . . . . . . . . . . . . . . . . . . . . . . . 9<br />

Taking Blood Samples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9<br />

Erythrocyte Count . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10<br />

Hemoglobin and Hematocrit Assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10<br />

Calculation <strong>of</strong> Erythrocyte Parameters . . . . . . . . . . . . . . . . . . . . . . . . . . 10<br />

Red Cell Distribution Width (RDW) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11<br />

Reticulocyte Count . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11<br />

Leukocyte Count . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14<br />

Thrombocyte Count . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15<br />

Quantitative Normal Values and Distribution <strong>of</strong> Cellular Blood<br />

Components . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15<br />

The Blood Smear and Its Interpretation (Differential Blood Count,<br />

DBC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17<br />

Significance <strong>of</strong> the Automated Blood Count . . . . . . . . . . . . . . . . . . . . . 19<br />

Bone Marrow Biopsy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20<br />

Lymph Node Biopsy and Tumor Biopsy . . . . . . . . . . . . . . . . . . . . . . . . . 23<br />

Step-by-Step Diagnostic Sequence . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs . 29<br />

The Individual Cells <strong>of</strong> Hematopoiesis . . . . . . . . . . . . . . . . . . . . . . . . 30<br />

Immature Red Cell Precursors: Proerythroblasts and Basophilic<br />

Erythroblasts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30<br />

Mature Red Blood Precursor Cells: Polychromatic and Orthochromatic<br />

Erythroblasts (Normoblasts) and Reticulocytes . . . . . . . 32<br />

Immature White Cell Precursors: Myeloblasts and Promyelocytes<br />

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


viii<br />

Contents<br />

Partly Mature White Cell Precursors: Myelocytes and Metamyelocytes<br />

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36<br />

Mature Neutrophils: Band Cells and Segmented Neutrophils . . . . . 38<br />

Cell Degradation, Special Granulations, and Nuclear Appendages<br />

in Neutrophilic Granulocytes and Nuclear Anomalies . . . . . . . . . . . . 40<br />

Eosinophilic Granulocytes (Eosinophils) . . . . . . . . . . . . . . . . . . . . . . . . 44<br />

Basophilic Granulocytes (Basophils) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44<br />

Monocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46<br />

Lymphocytes (and Plasma Cells) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48<br />

Megakaryocytes and Thrombocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50<br />

Bone Marrow: Cell Composition and Principles <strong>of</strong> Analysis . . . . 52<br />

Bone Marrow: Medullary Stroma Cells . . . . . . . . . . . . . . . . . . . . . . . . . 58<br />

Abnormalities <strong>of</strong> the White Cell Series . . . . . . . . . . . . . . . . . . 61<br />

Predominance <strong>of</strong> Mononuclear Round to Oval Cells . . . . . . . . . . . 63<br />

Reactive Lymphocytosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66<br />

Examples <strong>of</strong> Extreme Lymphocytic Stimulation: Infectious<br />

Mononucleosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68<br />

Diseases <strong>of</strong> the Lymphatic System (Non-Hodgkin Lymphomas) . . . 70<br />

Differentiation <strong>of</strong> the Lymphatic Cells and Cell Surface Marker<br />

Expression in Non-Hodgkin Lymphoma Cells . . . . . . . . . . . . . . . . . 72<br />

Chronic Lymphocytic Leukemia (CLL) and Related Diseases . . . . 74<br />

Lymphoplasmacytic Lymphoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78<br />

Facultative Leukemic Lymphomas (e.g., Mantle Cell Lymphoma<br />

and Follicular Lymphoma) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78<br />

Lymphoma, Usually with Splenomegaly (e.g., Hairy Cell Leukemia<br />

and Splenic Lymphoma with Villous Lymphocytes) . . . . . . . 80<br />

Monoclonal Gammopathy (Hypergammaglobulinemia), Multiple<br />

Myeloma*, Plasma Cell Myeloma, Plasmacytoma . . . . . . . . . 82<br />

Variability <strong>of</strong> Plasmacytoma Morphology . . . . . . . . . . . . . . . . . . . . . 84<br />

Relative Lymphocytosis Associated with Granulocytopenia<br />

(Neutropenia) and Agranulocytosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86<br />

Classification <strong>of</strong> Neutropenias and Agranulocytoses . . . . . . . . . . . 86<br />

Monocytosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88<br />

Acute Leukemias . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90<br />

Morphological and Cytochemical Cell Identification . . . . . . . . . . . 91<br />

Acute Myeloid Leukemias (AML) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95<br />

Acute Erythroleukemia (FAB Classification Type M6) . . . . . . . . . . 100<br />

Acute Megakaryoblastic Leukemia<br />

(FAB Classification Type M7) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102<br />

AML with Dysplasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102<br />

Hypoplastic AML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Contents<br />

Acute Lymphoblastic Leukemia (ALL) . . . . . . . . . . . . . . . . . . . . . . . . 104<br />

Myelodysplasia (MDS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106<br />

Prevalence <strong>of</strong> Polynuclear (Segmented) Cells . . . . . . . . . . . . . . . . . 110<br />

Neutrophilia without Left Shift . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110<br />

Reactive Left Shift . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112<br />

Chronic Myeloid Leukemia and Myeloproliferative Syndrome<br />

(Chronic Myeloproliferative Disorders, CMPD) . . . . . . . . . . . . . . . . . . 114<br />

Steps in the Diagnosis <strong>of</strong> Chronic Myeloid Leukemia . . . . . . . . . . 116<br />

Blast Crisis in Chronic Myeloid Leukemia . . . . . . . . . . . . . . . . . . . . . 120<br />

Osteomyelosclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122<br />

Elevated Eosinophil and Basophil Counts . . . . . . . . . . . . . . . . . . . . . . . 124<br />

Erythrocyte and Thrombocyte Abnormalities . . . . . . . . . . . 127<br />

Clinically Relevant Classification Principle for Anemias: Mean<br />

Erythrocyte Hemoglobin Content (MCH) . . . . . . . . . . . . . . . . . . . . . . . 128<br />

Hypochromic Anemias . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128<br />

Iron Deficiency Anemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128<br />

Hypochromic Infectious or Toxic Anemia (Secondary Anemia) . . . 134<br />

Bone Marrow Cytology in the Diagnosis <strong>of</strong> Hypochromic Anemias<br />

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136<br />

Hypochromic Sideroachrestic Anemias (Sometimes Normochromic<br />

or Hyperchromic) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137<br />

Hypochromic Anemia with Hemolysis . . . . . . . . . . . . . . . . . . . . . . . . . 138<br />

Thalassemias . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138<br />

Normochromic Anemias . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140<br />

Normochromic Hemolytic Anemias . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140<br />

Hemolytic Anemias with Erythrocyte Anomalies . . . . . . . . . . . . . . . . 144<br />

Normochromic Renal Anemia (Sometimes Hypochromic or<br />

Hyperchromic) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146<br />

Bone Marrow Aplasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146<br />

Pure Red Cell Aplasia (PRCA, Erythroblastopenia) . . . . . . . . . . . . . 146<br />

Aplasias <strong>of</strong> All Bone Marrow Series (Panmyelopathy, Panmyelophthisis,<br />

Aplastic Anemia) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 148<br />

Bone Marrow Carcinosis and Other Space-Occupying Processes . . 150<br />

Hyperchromic Anemias . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152<br />

Erythrocyte Inclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 156<br />

Hematological Diagnosis <strong>of</strong> Malaria . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

ix


x<br />

Contents<br />

Polycythemia Vera (Erythremic Polycythemia)<br />

and Erythrocytosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 162<br />

Thrombocyte Abnormalities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164<br />

Thrombocytopenia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164<br />

Thrombocytopenias Due to Increased Demand<br />

(High Turnover) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164<br />

Thrombocytopenias Due to Reduced Cell Production . . . . . . . . . . 168<br />

Thrombocytosis (Including Essential Thrombocythemia) . . . . . . . . 170<br />

Essential Thrombocythemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170<br />

Cytology <strong>of</strong> Organ Biopsies and Exudates . . . . . . . . . . . . . . . 173<br />

Lymph Node Cytology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174<br />

Reactive Lymph Node Hyperplasia and Lymphogranulomatosis<br />

(Hodgkin Disease) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 176<br />

Sarcoidosis and Tuberculosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 180<br />

Non-Hodgkin Lymphoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182<br />

Metastases <strong>of</strong> Solid Tumors in Lymph Nodes or Subcutaneous<br />

Tissue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182<br />

Branchial Cysts and Bronchoalveolar Lavage . . . . . . . . . . . . . . . . . . 184<br />

Branchial Cysts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184<br />

Cytology <strong>of</strong> the Respiratory System, Especially Bronchoalveolar<br />

Lavage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184<br />

Cytology <strong>of</strong> Pleural Effusions and Ascites . . . . . . . . . . . . . . . . . . . . . 186<br />

Cytology <strong>of</strong> Cerebrospinal Fluid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188<br />

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190<br />

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Physiology and Pathophysiology <strong>of</strong><br />

Blood Cells: Methods and Test<br />

Procedures<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


2<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

Introduction to the Physiology and<br />

Pathophysiology <strong>of</strong> the Hematopoietic<br />

System<br />

The reason why quantitative<br />

and qualitative diagnosis<br />

based on the cellular<br />

components <strong>of</strong> the blood is<br />

so important is that blood<br />

cells are easily accessible<br />

indicators <strong>of</strong> disturbances<br />

in their organs <strong>of</strong> origin or<br />

degradation—which are<br />

much less easily accessible.<br />

Thus, disturbances in the<br />

erythrocyte, granulocyte,<br />

and thrombocyte series<br />

allow important conclusions<br />

to be drawn about<br />

bone marrow function, just<br />

as disturbances <strong>of</strong> the lymphatic<br />

cells indicate reactions<br />

or disease states <strong>of</strong><br />

the specialized lymphopoietic<br />

organs (basically,<br />

the lymph nodes, spleen,<br />

and the diffuse lymphatic<br />

intestinal organ).<br />

Cell Systems<br />

All blood cells derive from a<br />

common stem cell. Under<br />

the influences <strong>of</strong> local and<br />

humoral factors, stem cells<br />

differentiate into different<br />

Fig. 1 Model <strong>of</strong> cell lineages <br />

in hematopoiesis<br />

NK cells<br />

NK cells<br />

Pluripotent<br />

lymphatic<br />

stem cells<br />

T-lymphopoiesis<br />

T-lymphoblasts<br />

T-lymphocytes<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

Pluripotent hemato-<br />

B-lymphopoiesis<br />

B-lymphoblasts<br />

B-lymphocytes<br />

Plasma cells


Omnipotent<br />

stem cells<br />

poietic stem cells<br />

Basophils<br />

Eosinophils<br />

Basophilic Eosinophilic<br />

promyelocytes promyelocytes<br />

Basophilic<br />

segmented<br />

granulocytes<br />

Eosinophilic<br />

segmented<br />

granulocytes<br />

Introduction to the Physiology and Pathophysiology<br />

cell lines (Fig. 1). Erythropoiesis and thrombopoiesis proceed independently<br />

once the stem cell stage has been passed, whereas monocytopoiesis<br />

and granulocytopoiesis are quite closely “related.” Lymphocytopoiesis<br />

is the most independent among the remaining cell series. Granulocytes,<br />

monocytes, and lymphocytes are collectively called leukocytes (white<br />

blood cells), a term that has been retained since the days before staining<br />

Monopoiesis<br />

Monoblasts<br />

Promonocytes<br />

Monocytes<br />

Macrophages<br />

Granulopoiesis<br />

monopoiesis<br />

Pluripotent myeloid<br />

stem cells<br />

Granulopoiesis<br />

Myeloblasts<br />

Promyelocytes<br />

Myelocytes<br />

Metamyelocytes<br />

Cells with band nuclei<br />

Neutrophilic<br />

granulocytes<br />

with segmented<br />

nuclei<br />

Thrombopoiesis<br />

Megakaryoblasts<br />

Megakaryocytes<br />

Thrombocytes<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

Erythropoiesis<br />

Proerythroblasts<br />

Erythroblasts<br />

Erythrocytes<br />

3


4<br />

a<br />

b<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

methods were available, when the only distinction that could be made<br />

was between erythrocytes (red blood cells) and the rest.<br />

All these cells are eukaryotic, that is, they are made up <strong>of</strong> a nucleus,<br />

sometimes with visible nucleoli, surrounded by cytoplasm, which may include<br />

various kinds <strong>of</strong> organelles, granulations, and vacuoles.<br />

Despite the common origin <strong>of</strong> all the cells, ordinary light microscopy<br />

reveals fundamental and characteristic differences in the nuclear chromatin<br />

structure in the different cell series and their various stages <strong>of</strong><br />

maturation (Fig. 2).<br />

The developing cells in the granulocyte series (myeloblasts and promyelocytes),<br />

for example, show a delicate, fine “net-like” (reticular) structure.<br />

Careful microscopic examination (using fine focus adjustment to<br />

view different depth levels) reveals a detailed nuclear structure that resembles<br />

fine or coarse gravel (Fig. 2 a). With progressive stages <strong>of</strong> nuclear<br />

maturation in this series (myelocytes, metamyelocytes, and band or staff<br />

cells), the chromatin condenses into bands or streaks, giving the nucleus—<br />

which at the same time is adopting a characteristic curved shape—a<br />

spotted and striped pattern (Fig. 2 b).<br />

Lymphocytes, on the other hand—particularly in their circulating<br />

forms—always have large, solid-looking nuclei. Like cross-sections<br />

through geological slate, homogeneous, dense chromatin bands alternate<br />

with lighter interruptions and fissures (Fig. 2 c).<br />

Each <strong>of</strong> these cell series contains precursors that can divide (blast precursors)<br />

and mature or almost mature forms that can no longer divide; the<br />

morphological differences between these correspond not to steps in mito-<br />

Vacuoles Nucleus (with<br />

delicate reticular<br />

chromatin structure)<br />

Lobed nucleus with<br />

banded chromatin<br />

structure<br />

Cytoplasm<br />

Nucleolus<br />

Cytoplasmic granules<br />

c<br />

Coarse chromatin<br />

structure<br />

Fig. 2 Principles <strong>of</strong><br />

cell structure with examples<br />

<strong>of</strong> different<br />

nuclear chromatin<br />

structure. a Cell <strong>of</strong> the<br />

myeloblast to promyelocyte<br />

type. b Cell<br />

<strong>of</strong> the myelocyte to<br />

staff or band cell type.<br />

c Cell <strong>of</strong> the lymphocyte<br />

type with<br />

coarsely structured<br />

chromatin<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Introduction to the Physiology and Pathophysiology<br />

sis, but result from continuous “maturation processes” <strong>of</strong> the cell nucleus<br />

and cytoplasm. Once this is understood, it becomes easier not to be too<br />

rigid about morphological distinctions between certain cell stages. The<br />

blastic precursors usually reside in the hematopoietic organs (bone marrow<br />

and lymph nodes). Since, however, a strict blood–bone marrow barrier<br />

does not exist (blasts are kept out <strong>of</strong> the bloodstream essentially only<br />

by their limited plasticity, i.e., their inability to cross the diffusion barrier<br />

into the bloodstream), it is in principle possible for any cell type to be<br />

found in peripheral blood, and when cell production is increased, the<br />

statistical frequency with which they cross into the bloodstream will naturally<br />

rise as well. Conventionally, cells are sorted left to right from immature<br />

to mature, so an increased level <strong>of</strong> immature cells in the bloodstream<br />

causes a “left shift” in the composition <strong>of</strong> a cell series—although it<br />

must be said that only in the precursor stages <strong>of</strong> granulopoiesis are the cell<br />

morphologies sufficiently distinct for this left shift to show up clearly.<br />

The distribution <strong>of</strong> white blood cells outside their places <strong>of</strong> origin cannot<br />

be inferred simply from a drop <strong>of</strong> capillary blood. This is because the<br />

majority <strong>of</strong> white cells remain out <strong>of</strong> circulation, “marginated” in the<br />

epithelial lining <strong>of</strong> vessel walls or in extravascular spaces, from where<br />

they may be quickly recruited back to the bloodstream. This phenomenon<br />

explains why white cell counts can vary rapidly without or before any<br />

change has taken place in the rate <strong>of</strong> their production.<br />

Cell functions. A brief indication <strong>of</strong> the functions <strong>of</strong> the various cell groups<br />

follows (see Table 1).<br />

Neutrophil granulocytes with segmented nuclei serve mostly to<br />

defend against bacteria. Predominantly outside the vascular system, in “inflamed”<br />

tissue, they phagocytose and lyse bacteria. The blood merely<br />

transports the granulocytes to their site <strong>of</strong> action.<br />

The function <strong>of</strong> eosinophilic granulocytes is defense against parasites;<br />

they have a direct cytotoxic action on parasites and their eggs and larvae.<br />

They also play a role in the down-regulation <strong>of</strong> anaphylactic shock reactions<br />

and autoimmune responses, thus controlling the influence <strong>of</strong> basophilic<br />

cells.<br />

The main function <strong>of</strong> basophilic granulocytes and their tissue-bound<br />

equivalents (tissue mast cells) is to regulate circulation through the release<br />

<strong>of</strong> substances such as histamine, serotonin, and heparin. These tissue<br />

hormones increase vascular permeability at the site <strong>of</strong> various local antigen<br />

activity and thus regulate the influx <strong>of</strong> the other inflammatory cells.<br />

The main function <strong>of</strong> monocytes is the defense against bacteria, fungi,<br />

viruses, and foreign bodies. Defensive activities take place mostly outside<br />

the vessels by phagocytosis. Monocytes also break down endogenous cells<br />

(e.g., erythrocytes) at the end <strong>of</strong> their life cycles, and they are assumed to<br />

perform a similar function in defense against tumors. Outside the bloodstream,<br />

monocytes develop into histiocytes; macrophages in the<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

5


6<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

endothelium <strong>of</strong> the body cavities; epithelioid cells; foreign body macrophages<br />

(including Langhans’ giant cells); and many other cells.<br />

Lymphocytes are divided into two major basic groups according to<br />

function.<br />

Thymus-dependent T-lymphocytes, which make up about 70% <strong>of</strong><br />

lymphocytes, provide local defense against antigens from organic and inorganic<br />

foreign bodies in the form <strong>of</strong> delayed-type hypersensitivity, as classically<br />

exemplified by the tuberculin reaction. T-lymphocytes are divided<br />

into helper cells and suppressor cells. The small group <strong>of</strong> NK (natural<br />

killer) cells, which have a direct cytotoxic function, is closely related to the<br />

T-cell group.<br />

The other group is the bone-marrow-dependent B-lymphocytes or Bcells,<br />

which make up about 20% <strong>of</strong> lymphocytes. Through their development<br />

into immunoglobulin-secreting plasma cells, B-lymphocytes are responsible<br />

for the entire humoral side <strong>of</strong> defense against viruses, bacteria,<br />

and allergens.<br />

Table 1 Cells in a normal peripheral blood smear and their physiological roles<br />

Cell type Function Count<br />

(% <strong>of</strong> leukocytes)<br />

Neutrophilic band<br />

granulocytes (band<br />

neutrophil)<br />

Neutrophilic segmented<br />

granulocyte (segmented<br />

neutrophil)<br />

Lymphocytes<br />

(B- and T-lymphocytes,<br />

morphologically indistinguishable)<br />

Precursors <strong>of</strong> segmented cells<br />

that provide antibacterial<br />

immune response<br />

Phagocytosis <strong>of</strong> bacteria;<br />

migrate into tissue for this purpose<br />

B-lymphocytes (20% <strong>of</strong> ⎫<br />

lymphocytes) mature and ⎪<br />

form plasma cells antibody<br />

⎪<br />

⎪<br />

production.<br />

⎬<br />

T-lymphocytes (70%): cyto- ⎪<br />

toxic defense against viruses, ⎪<br />

foreign antigens, and tumors. ⎭<br />

Monocytes Phagocytosis <strong>of</strong> bacteria, protozoa,<br />

fungi, foreign bodies.<br />

Transformation in target tissue<br />

Eosinophilic granulocytes Immune defense against parasites,<br />

immune regulation<br />

Basophilic granulocytes Regulation <strong>of</strong> the response to<br />

local inflammatory processes<br />

0–4%<br />

50–70%<br />

20–50%<br />

2–8%<br />

1–4%<br />

0–1%<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Introduction to the Physiology and Pathophysiology<br />

Erythrocytes are the oxygen carriers for all oxygen-dependent metabolic<br />

reactions in the organism. They are the only blood cells without nuclei,<br />

since this allows them to bind and exchange the greatest number <strong>of</strong><br />

O2 molecules. Their physiological biconcave disk shape with a thick rim<br />

provides optimal plasticity.<br />

Thrombocytes form the aggregates that, along with humoral coagulation<br />

factors, close up vascular lesions. During the aggregation process, in<br />

addition to the mechanical function, thrombocytic granules also release<br />

factors that promote coagulation.<br />

Thrombocytes develop from polyploid megakaryocytes in the bone<br />

marrow. They are the enucleated, fragmented cytoplasmic portions <strong>of</strong><br />

these progenitor cells.<br />

Principles <strong>of</strong> Regulation and Dysregulation in the<br />

Blood Cell Series and their Diagnostic Implications<br />

Quantitative and qualitative equilibrium between all blood cells is maintained<br />

under normal conditions through regulation by humoral factors,<br />

which ensure a balance between cell production (mostly in the bone marrow)<br />

and cell degradation (mostly in the spleen, liver, bone marrow, and<br />

the diffuse reticular tissue).<br />

Compensatory increases in cell production are induced by cell loss or increased<br />

cell demand. This compensatory process can lead to qualitative<br />

changes in the composition <strong>of</strong> the blood, e.g., the occurrence <strong>of</strong> nucleated<br />

red cell precursors compensating for blood loss or increased oxygen requirement,<br />

or following deficiency <strong>of</strong> certain metabolites (in the restitution<br />

phase, e.g., during iron or vitamin supplementation). Similarly,<br />

during acute immune reactions, which lead to an increased demand for<br />

cells, immature leukocyte forms may appear (“left shift”).<br />

Increased cell counts in one series can lead to suppression <strong>of</strong> cell production<br />

in another series. The classic example is the suppression <strong>of</strong> erythrocyte<br />

production (the pathomechanical details <strong>of</strong> which are incompletely<br />

understood) during infectious/toxic reactions, which affect the white cells<br />

(“infectious anemia”).<br />

Metabolite deficiency as a pathogenic stimulus affects the erythrocyte series<br />

first and most frequently. Although other cell series are also affected,<br />

this series, with its high turnover, is the one most vulnerable to metabolite<br />

deficiencies. Iron deficiency, for example, rapidly leads to reduced<br />

hemoglobin in the erythrocytes, while vitamin B12 and/or folic acid deficiency<br />

will result in complex disturbances in cell formation. Eventually,<br />

these disturbances will start to show effects in the other cell series as well.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

7


8<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

Toxic influences on cell production usually affect all cell series. The effects<br />

<strong>of</strong> toxic chemicals (including alcohol), irradiation, chronic infections, or<br />

tumor load, for example, usually lead to a greater or lesser degree <strong>of</strong> suppression<br />

in all the blood cell series, lymphocytes and thrombocytes being<br />

the most resistant. The most extreme result <strong>of</strong> toxic effects is panmyelophthisis<br />

(the synonym “aplastic anemia” ignores the fact that the leukocyte<br />

and thrombocyte series are usually also affected).<br />

Autoimmune and allergic processes may selectively affect a single cell series.<br />

Results <strong>of</strong> this include “allergic” agranulocytosis, immunohemolytic<br />

anemia, and thrombocytopenia triggered by either infection or medication.<br />

Autoimmune suppression <strong>of</strong> the pluripotent stem cells can also<br />

occur, causing panmyelophthisis.<br />

Malignant dedifferentiation can basically occur in cells <strong>of</strong> any lineage at<br />

any stage where the cells are able to divide, causing chronic or acute clinical<br />

manifestations. These deviations from normal differentiation occur most<br />

frequently in the white cell series, causing “leukemias.” Recent data indicate<br />

that in fact in these cases the remaining cell series also become distorted,<br />

perhaps via generalized atypical stem cell formation. Erythroblastosis,<br />

polycythemia, and essential thrombocythemia are examples showing<br />

that malignant processes can also manifest themselves primarily in<br />

the erythrocyte or thrombocyte series.<br />

Malignant “transformations” always affect blood cell precursors that<br />

are still capable <strong>of</strong> dividing, and the result is an accumulation <strong>of</strong> identical,<br />

constantly self-reproducing blastocytes. These are not necessarily always<br />

observed in the bloodstream, but can remain in the bone marrow. That is<br />

why, in “leukemia,” it is <strong>of</strong>ten not the number <strong>of</strong> cells, but the increasing<br />

lack <strong>of</strong> normal cells that is the indicative hematological finding.<br />

All disturbances <strong>of</strong> bone marrow function are accompanied by quantitative<br />

and/or qualitative changes in the composition <strong>of</strong> blood cells or<br />

blood proteins. Consequently, in most disorders, careful analysis <strong>of</strong><br />

changes in the blood together with clinical findings and other laboratory<br />

data produces the same information as bone marrow cytology. The relationship<br />

between the production site (bone marrow) and the destination<br />

(the blood) is rarely so fundamentally disturbed that hematological analysis<br />

and humoral parameters will not suffice for a diagnosis. This is virtually<br />

always true for hypoplastic–anaplastic processes in one or all cell series<br />

with resulting cytopenia but without hematological signs <strong>of</strong> malignant<br />

cell proliferation.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Procedures, Assays, and Normal Values<br />

Taking Blood Samples<br />

Procedures, Assays, and Normal Values<br />

Since cell counts are affected by the state <strong>of</strong> the blood circulation, the<br />

conditions under which samples are taken should be the same so far as<br />

possible if comparable values are desired.<br />

This means that blood should always be drawn at about the same time <strong>of</strong><br />

day and after at least eight hours <strong>of</strong> fasting, since both circadian rhythm<br />

and nutritional status can affect the findings. If strictly comparable values<br />

are required, there should also be half an hour <strong>of</strong> bed rest before the<br />

sample is drawn, but this is only practicable in a hospital setting. In other<br />

settings (i.e., outpatient clinics), bringing portable instruments to the relaxed,<br />

seated patient works well.<br />

A sample <strong>of</strong> capillary blood may be taken when there are no further<br />

tests that would require venous access for a larger sample volume. A wellperfused<br />

fingertip or an earlobe is ideal; in newborns or young infants, the<br />

heel is also a good site. If the circulation is poor, the blood flow can be increased<br />

by warming the extremity by immersing it in warm water.<br />

Without pressure, the puncture area is swabbed several times with 70%<br />

alcohol, and the skin is then punctured firmly but gently with a sterile disposable<br />

lancet. The first droplet <strong>of</strong> blood is discarded because it may be<br />

contaminated, and the ensuing blood is drawn into the pipette (see<br />

below). Care should be taken not to exert pressure on the tissue from<br />

which the blood is being drawn, because this too can change the cell composition<br />

<strong>of</strong> the sample.<br />

Obviously, if a venous blood sample is to be taken for the purposes <strong>of</strong><br />

other tests, or if an intravenous injection is going to be performed, the<br />

blood sample for hematological analysis can be taken from the same site.<br />

To do this, the blood is allowed to flow via an intravenous needle into a<br />

specially prepared (commercially available) EDTA-treated tube. The tube<br />

is filled to the 1-ml mark and then carefully shaken several times. The very<br />

small amount <strong>of</strong> EDTA in the tube prevents the blood from clotting, but<br />

can itself be safely ignored in the quantitative analysis.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

9


10<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

Erythrocyte Count<br />

Up to 20 years ago, blood cells were counted “by hand” in an optical counting<br />

chamber. This method has now been almost completely abandoned in<br />

favor <strong>of</strong> automated counters that determine the number <strong>of</strong> erythrocytes<br />

by measuring the impedance or light dispersion <strong>of</strong> EDTA blood (1 ml), or<br />

heparinized capillary blood. Due to differences in the hematocrit, the<br />

value from a sample taken after (at least 15 minutes’) standing or physical<br />

activity will be 5–10% higher than the value from a sample taken after 15<br />

minutes’ bed rest.<br />

Hemoglobin and Hematocrit Assay<br />

Hemoglobin is oxidized to cyanmethemoglobin by the addition <strong>of</strong> cyanide,<br />

and the cyanmethemoglobin is then determined spectrophotometrically<br />

by the automated counter. The hematocrit describes the ratio<br />

<strong>of</strong> the volume <strong>of</strong> erythrocytes to the total blood volume (the SI unit is<br />

without dimension, e.g., 0.4).<br />

The EDTA blood is centrifuged in a disposable capillary tube for 10<br />

minutes using a high-speed microhematocrit centrifuge (reference<br />

method). The automated hematology counter determines the mean corpuscular<br />

or cell volume (MCV, measured in femtoliters, fl) and the number<br />

<strong>of</strong> erythrocytes. It calculates the hematocrit (HCT) using the following<br />

formula:<br />

HCT = MCV (fl) number <strong>of</strong> erythrocytes (10 6 /µl).<br />

Calculation <strong>of</strong> Erythrocyte Parameters<br />

The quality <strong>of</strong> erythrocytes is characterized by their MCV, their mean cell<br />

hemoglobin content (MCH), and the mean cellular hemoglobin concentration<br />

(MCHC).<br />

MCV is measured directly using an automated hemoglobin analyzer, or<br />

is calculated as follows:<br />

MCV <br />

Hematocrit (l/l)<br />

Number <strong>of</strong> erythrocytes (10 6 /µl)<br />

MCH (in picograms per erythrocyte) is calculated using the following<br />

formula:<br />

Hemoglobin (g/l)<br />

MCH (pg) <br />

Number <strong>of</strong> erythrocytes (106 /µl)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


MCHC is determined using this formula:<br />

MCHC (g/dl) <br />

Hemoglobin concentration (g/dl)<br />

Hematocrit (l/l)<br />

Red Cell Distribution Width (RDW)<br />

Modern analyzers also record the red cell distribution width (cell volume<br />

distribution). In normal erythrocyte morphology, this correlates with the<br />

Price-Jones curve for the cell diameter distribution. Discrepancies are<br />

used diagnostically and indicate the presence <strong>of</strong> microspherocytes<br />

(smaller cells with lighter central pallor).<br />

Reticulocyte Count<br />

Procedures, Assays, and Normal Values<br />

Reticulocytes can be counted using flow cytometry and is based on the<br />

light absorbed by stained aggregates <strong>of</strong> reticulocyte organelles. The data<br />

are recorded as the number <strong>of</strong> reticulocytes per mill (‰) <strong>of</strong> the total number<br />

<strong>of</strong> erythrocytes. Reticulocytes can, <strong>of</strong> course, be counted in a counting<br />

chamber using a microscope. While this method is not particularly<br />

laborious, it is mostly employed in laboratories that <strong>of</strong>ten deal with or<br />

have a special interest in anemia. Reticulocytes are young erythrocytes<br />

immediately after they have extruded their nuclei: they contain, as a remainder<br />

<strong>of</strong> aggregated cell organelles, a net-like structure (hence the<br />

name “reticulocyte”) that is not discernible after the usual staining procedures<br />

for leukocytes, but can be observed after vital staining <strong>of</strong> cells<br />

with brilliant cresyl blue or new methylene blue. The staining solution is<br />

mixed in an Eppendorf tube with an equal volume <strong>of</strong> EDTA blood and incubated<br />

for 30 minutes. After repeated mixing, a blood smear is prepared<br />

and allowed to dry. The sample is viewed using a microscope equipped<br />

with an oil immersion lens. The ratio <strong>of</strong> reticulocytes to erythrocytes is determined<br />

and plotted as reticulocytes per 1000 erythrocytes (per mill).<br />

Normal values are listed in Table 2, p. 12.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

11


12<br />

Table 2 Normal ranges and mean values for blood cell components*<br />

Adults Newborns Toddlers Children<br />

18 years old 1 months 2 years old 10 years old<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

MV 7000 11000 10000 8000<br />

NR 4300–10000<br />

Leukocytes/µl<br />

or 106 /l**<br />

Band granulocytes % MV 2 5 3 3<br />

NR 0–5<br />

Segmented neutrophilic MV 60 30 30 30<br />

granulocytes<br />

NR 35–85<br />

absolute ct./µl MV 3650 3800 3500 4400<br />

or 106 /l** NR 1850–7250<br />

Lymphocytes % MV 30 55 60 40<br />

NR 20–50<br />

absolute ct./µl MV 2500 6000 6300 3100<br />

or 106 /l** NR 1500–3500<br />

Monocytes % MV 4 6 5 4<br />

absolute ct./µl NR 2–6<br />

or 106 /l** MV 450<br />

NR 70–840<br />

Eosinophilic granulocytes (%) MV 2 3 2 2<br />

NR 0–4<br />

absolute ct./µl MV 165<br />

or 106 /l** NR 0–400<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Basophilic granulocytes (%) MW 0.5 0.5 0.5 0.5<br />

NR 0–1<br />

Male Female<br />

MV 5.4 4.8 4.7 4.7 4.8<br />

NR 4.6–5.9 4.2–5.4 3.9–5.9 3.8–5.4 3.8–5.4<br />

Erythrocytes 106 /µl<br />

or 1012 /l**<br />

MV 15 13 17 12 14<br />

NR 14–18 12–16 15–18 11–13 12–15<br />

Hb g/dl<br />

or 10 g/l**<br />

HKT MV 0.45 0.42 44 37 39<br />

NR 0.42–0.48 0.38–0.43<br />

Procedures, Assays, and Normal Values<br />

MCH HbE (pg) MV 29 33 27 25<br />

NR 26–32<br />

MV 87 91 78 80<br />

NR 77–99<br />

MCV/µm3 or fl**<br />

MV 33 35 33 34<br />

NR 33–36<br />

MCHC g/dl<br />

or 10 g/l<br />

Erythrocyte, diameter (µm) MV 7.5 8.1 7.3 7.4<br />

Reticulocytes (%) MV 16 24 7.9 7.1 7.6<br />

NR 8–25 8–40<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

Thrombocytes 103 /µl MV 180 155–566 286–509 247–436<br />

NR 140–440<br />

13<br />

MV mean value, NR normal range (range for 95% <strong>of</strong> the population, reference range), ct. count, ** SI units give the measurements per liter.<br />

* For technical reasons, data may vary considerably between laboratories. It is therefore important also to consult the reference ranges <strong>of</strong> the chosen laboratory.


14<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

Leukocyte Count<br />

Leukocytes, unlike erythrocytes, are completely colorless in their native<br />

state. Another important physical difference is the stability <strong>of</strong> leukocytes<br />

in 3% acetic acid or saponins; these media hemolyze erythrocytes (though<br />

not their nucleated precursors). Türk’s solution, used in most counting<br />

methods, employs glacial acetic acid for hemolysis and crystal violet (gentian<br />

violet) to lightly stain the leukocytes. A 50-µl EDTA blood sample is<br />

mixed with 500 µl Türk’s solution in an Eppendorf tube and incubated at<br />

room temperature for 10 minutes. The suspension is again mixed and<br />

carefully transferred to the well <strong>of</strong> a prepared counting chamber using a<br />

pipette or capillary tube. The chamber is allowed to fill from a droplet<br />

placed at one edge <strong>of</strong> the well and placed in a moisture-saturated incubator<br />

for 10 minutes. With the condenser lowered (or using phase contrast<br />

microscopy), the leukocytes are then counted in a total <strong>of</strong> four <strong>of</strong> the large<br />

squares opposite to each other (1 mm 2 each). The result is multiplied by<br />

27.5 (dilution: 1 + 10, volume: 0.4 mm 3 ) to yield the leukocyte count per<br />

microliter. Parallel (control) counts show variation <strong>of</strong> up to 15%. The normal<br />

(reference) ranges are given in Table 2.<br />

In an automated blood cell counter, the erythrocytes are lysed and cells<br />

with a volume that exceeds about 30 fl (threshold values vary for different<br />

instruments) are counted as leukocytes. Any remaining erythroblasts,<br />

hard-to-lyse erythrocytes such as target cells, giant thrombocytes, or agglutinated<br />

thrombocytes are counted along with the leukocytes, and this<br />

will lead to an overestimate <strong>of</strong> the leukocyte count. Modern analyzers can<br />

recognize such interference factors and apply interference algorithms to<br />

obtain a corrected leukocyte count.<br />

Visual leukocyte counts using a counting chamber show a variance <strong>of</strong><br />

about 10%; they can be used as a control reference for automatic cell<br />

counts. Rough estimates can also be made by visual assessment <strong>of</strong> blood<br />

smears: a 40 objective will show an average <strong>of</strong> two to three cells per field<br />

<strong>of</strong> view if the leukocyte count is normal.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Thrombocyte Count<br />

Procedures, Assays, and Normal Values<br />

15<br />

To count thrombocytes in a counting chamber, blood must be conditioned<br />

with 2% Novocain–Cl solution. Preprepared commercial tubes are widely<br />

used (e.g., Thrombo Plus with 2-ml content). EDTA blood is pipetted into<br />

the tubes, carefully mixed and immediately placed in a counting chamber.<br />

The chamber is allowed to stand for 10 minutes while the cells settle,<br />

after which an area <strong>of</strong> 1 mm 2 is counted. The result corresponds to the<br />

number <strong>of</strong> thrombocytes (the 1 + 100 dilution is ignored). In an automated<br />

blood cell counter, the blood cells are counted after they have been sorted<br />

by size. Small cells between 2 and 20 fl (thresholds vary for different instruments)<br />

are counted as thrombocytes. If giant thrombocytes or agglutinated<br />

thrombocytes are present, they are not counted and the result is an<br />

underestimate. On the other hand, small particles, such as fragmentocytes,<br />

or microcytes, will lead to an overestimate. Modern analyzers can<br />

recognize such interference factors and apply interference algorithms to<br />

obtain a corrected thrombocyte count. If unexpected results are produced,<br />

it is wise to check them by direct reference to the blood smear.<br />

Unexpected thrombocyte counts should be verified by direct visual<br />

assessment. Using a 100 objective, the field <strong>of</strong> view normally contains<br />

an average <strong>of</strong> 10 thrombocytes. In some instances, “pseudothrombocytopenias”<br />

are found in automated counts. These are artifacts due to thrombocyte<br />

aggregation.<br />

Pseudothrombocytopenia (see p. 167) is caused by the aggregation <strong>of</strong><br />

thrombocytes in the presence <strong>of</strong> EDTA; it does not occur when heparin or<br />

citrate are used as anticoagulants.<br />

Quantitative Normal Values and Range <strong>of</strong><br />

Cellular Blood Components<br />

Determining normal values for blood components is more difficult and<br />

more risky than one might expect. Obviously, the values are affected by a<br />

large number <strong>of</strong> variables, such as age, gender, activity (metabolic load),<br />

circadian rhythm, and nutrition, not to mention the effects <strong>of</strong> the blood<br />

sampling technique, type and storage <strong>of</strong> the blood, and the counting<br />

method. For this reason, where available, a normal range is given, covering<br />

95% <strong>of</strong> the values found in a clinically normal group <strong>of</strong> probands—from<br />

which it follows that one in every 20 healthy people will have values outside<br />

the limits <strong>of</strong> this range. Thus, there are areas <strong>of</strong> overlap between normal<br />

and pathological data. Data in these borderline areas must be interpreted<br />

within a refined reference range with data from probands who<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


16<br />

Leukocyte count (10 9 /l)<br />

22<br />

20<br />

18<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

resemble each other and the patient as closely as possible in respect <strong>of</strong> the<br />

variables listed above. Due to space limitations, only key age data are considered<br />

here. Figure 3 clearly shows that, particularly for newborns, toddlers,<br />

and young children, particular reference ranges must be taken into<br />

account. In addition, the interpretation must also take account <strong>of</strong><br />

methodological variation: in cell counts, the coefficient <strong>of</strong> variation (standard<br />

deviation as a percentage <strong>of</strong> the mean value) is usually around 10!<br />

Hours<br />

12 2 4 6 1 2 3 5 3 6 9 1 3 5 7 9 11 13<br />

Days<br />

Weeks<br />

Months<br />

Years<br />

Leukocytes<br />

Granulocytes<br />

Lymphocytes<br />

Monocytes<br />

Fig. 3 Mean cell counts at different ages in childhood for leukocytes and their<br />

subfractions (according to Kato)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Procedures, Assays, and Normal Values<br />

In sum, a healthy distrust for the single data point is the most important<br />

basis for the interpretation <strong>of</strong> all data, including those outside the reference<br />

range. For every sample <strong>of</strong> drawn blood, and every counting<br />

method, at least two or three values should be available before conclusions<br />

can be drawn, unless the clinical findings reflect the cytological data.<br />

In addition to this, every laboratory has its own set <strong>of</strong> reference data to<br />

some extent.<br />

17<br />

After this account <strong>of</strong> the problems and wide variations between different<br />

groups, the data in Table 2 are presented in a simplified form, with values<br />

rounded up or down for ease <strong>of</strong> comparison and memorization. Absolute<br />

values and the new SI units are given where they are clinically relevant.<br />

The Blood Smear and Its Interpretation<br />

(Differential Blood Count, DBC)<br />

A blood smear uses capillary or venous EDTA-blood, preferably no more<br />

than three hours old. The slides must be grease-free, otherwise cell aggregation<br />

and stain precipitation may occur. Unless commercially available<br />

grease-free slides are used, the slides should be soaked for several hours in<br />

a solution <strong>of</strong> equal parts <strong>of</strong> ethanol and ether and then allowed to dry. A<br />

droplet <strong>of</strong> the blood sample is placed close to the edge <strong>of</strong> the slide. A<br />

ground cover glass (spreader slide) is placed in front <strong>of</strong> the droplet onto<br />

the slide at an angle <strong>of</strong> about 30. The cover slide is then slowly backed into<br />

the blood droplet. Upon contact, the blood droplet spreads along the edge<br />

<strong>of</strong> the slide (Fig. 4). Without pressure, the cover glass is now lightly moved<br />

over the slide. The faster the cover glass is moved, and the steeper angle at<br />

which it is held, the thinner the smear will be.<br />

The quality <strong>of</strong> the smear technique is crucial for the assessment, because<br />

the cell density at the end <strong>of</strong> the smear is <strong>of</strong>ten twice that at the beginning.<br />

In a well-prepared smear the blood sample will show a “feathered” edge<br />

where the cover glass left the surface <strong>of</strong> the slide. The smear must be<br />

thoroughly air-dried; for good staining, at least two hours’ drying time is<br />

needed. The quality <strong>of</strong> the preparation will be increased by 10 minutes’<br />

fixation with methanol, and it will then also keep better. After drying,<br />

name and date are pencilled in on the slide.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


18<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

Fig. 4 Preparation <strong>of</strong> a blood smear<br />

For safety’s sake, at least one back-up smear should be made from every<br />

sample from every patient.<br />

Staining is done with a mixture <strong>of</strong> basic stains (methylene blue, azure) and<br />

acidic stains (eosin), so as to show complementary substances such as nucleic<br />

acids and alkaline granulations. In addition to these leukocyte components,<br />

erythrocytes also yield different staining patterns: immature<br />

erythrocytes contain larger residual amounts <strong>of</strong> RNA and therefore stain<br />

more heavily with basophilic stains than do mature erythrocytes. Pappenheim’s<br />

panoptic stain contains a balanced mixture <strong>of</strong> basic and acidic<br />

stains: the horizontally stored, air-dried smear is covered with May–<br />

Grünwald staining solution (eosin–methylene blue) for three minutes,<br />

then about an equal amount <strong>of</strong> phosphate buffer, pH = 7.3, is carefully<br />

added and, after a further three minutes, carefully poured <strong>of</strong>f again. Next,<br />

the slide is covered with diluted Giemsa stain (azure–eosin), which is prepared<br />

by addition <strong>of</strong> 1 ml Giemsa stock solution to 1 ml neutral distilled<br />

water or phosphate buffer, pH = 6.8–7.4. After 15 minutes, the Giemsa<br />

staining solution is gently rinsed <strong>of</strong>f with buffer solution and the smears<br />

are air-dried with the feathered end sloping upwards.<br />

The blood smears are initially viewed with a smaller objective (10 to<br />

20), which allows the investigator to check the cell density and to find<br />

the best counting area in the smear. Experience shows that the cell projection<br />

is best about 1 cm from the feathered end <strong>of</strong> the smear. At 40 magnification,<br />

one may expect to see an average <strong>of</strong> two to three leukocytes per<br />

viewing field if the leukocyte count is normal. It is sometimes useful to be<br />

able to use this rough estimate to crosscheck improbable quantitative<br />

values. The detailed analysis <strong>of</strong> the white blood cells is done using an oil<br />

immersion lens and 100 magnification. For this, it is best to scan the sec-<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Procedures, Assays, and Normal Values<br />

19<br />

tion from about 1 cm to about 3 cm from the end <strong>of</strong> the smear, moving the<br />

slide to and fro in a meandering movement across its short diameter.<br />

Before (and while) the differential leukocyte count is carried out, erythrocyte<br />

morphology and thrombocyte density should be assessed. The results<br />

<strong>of</strong> the differential leukocyte count (the morphologies are presented in the<br />

atlas section, p. 30 ff.) may be recorded using manual counters or mark-up<br />

systems. The more cells are counted, the more representative the results,<br />

so when pathological deviations are found, it is advisable to count 200<br />

cells.<br />

To speed up the staining process, which can seem long and laborious<br />

when a rapid diagnosis is required, several quick-staining sets are available<br />

commercially, although most <strong>of</strong> them do not permit comparable fine<br />

analysis. If the standard staining solutions mentioned above are to hand, a<br />

quick stain for orientation purposes can be done by incubating the smear<br />

with May–Grünwald reagent for just one minute and shortening the<br />

Giemsa incubation time to one to two minutes with concentrated “solution.”<br />

Normal values and ranges for the differential blood count are given in<br />

Table 2, p. 12.<br />

Malaria plasmodia are best determined using a thick smear in addition<br />

to the normal blood smear. On a slide, a drop <strong>of</strong> blood is spread over an<br />

area <strong>of</strong> about 2.5 cm across. The thick smear is placed in an incubator and<br />

allowed to dry for at least 30 minutes. Drying samples as thick smears and<br />

then treating them with dilute Giemsa stain (as described above) achieves<br />

extensive hemolysis <strong>of</strong> the erythrocytes and thus an increase in the released<br />

plasmodia.<br />

Significance <strong>of</strong> the Automated Blood Count<br />

The qualitative and quantitative blood count techniques described here<br />

may seem somewhat archaic given the now almost ubiquitous automated<br />

cell counters; they are merely intended to show the possibilities always<br />

ready to be called on in terms <strong>of</strong> individual analyses carried out by small,<br />

dedicated laboratories.<br />

The automated cell count has certainly rationalized blood cell counting.<br />

Depending on the diagnostic problem and the quality control system <strong>of</strong><br />

the individual laboratory, automated counting can even reduce data<br />

ranges compared with “manual” counts.<br />

After lysis <strong>of</strong> the erythrocytes, hematology analyzers determine the number<br />

<strong>of</strong> remaining nucleated cells using a wide range <strong>of</strong> technologies. All<br />

counters use cell properties such as size, interaction with scattered light at<br />

different angles, electrical conductivity, nucleus-to-cytoplasm ratio, and<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


20<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

the peroxidase reaction, to group individual cell impulses into clusters.<br />

These clusters are then quantified and assigned to leukocyte populations.<br />

If only normal blood cells are present, the assignment <strong>of</strong> the clusters to<br />

the various leukocyte populations works well, and the precision <strong>of</strong> the automated<br />

count exceeds that <strong>of</strong> the manual count <strong>of</strong> 100 cells in a smear by<br />

a factor <strong>of</strong> 10. If large number <strong>of</strong> pathological cells are present, such as<br />

blasts or lymphadenoma cells, samples are reliably recognized as “pathological,”<br />

and a smear can then be prepared and further analyzed under the<br />

microscope.<br />

The difficulty arises when small populations <strong>of</strong> pathological cells are<br />

present (e.g., 2% blasts present after chemotherapy), or when pathological<br />

cells are present that closely resemble normal leukocytes (e.g., small centrocytes<br />

in satellite cell lymphoma). These pathological conditions are not<br />

always picked up by automated analyzers (false negative result), no smear<br />

is prepared and studied under the microscope, and the results produced<br />

by the machine do not include the presence <strong>of</strong> these pathological populations.<br />

For this reason, blood samples accompanied by appropriate clinical<br />

queries (e.g., “lymphadenoma?” “blasts?” “unexplained anemia?”) should<br />

always be differentiated and evaluated using a microscope.<br />

Bone Marrow Biopsy<br />

Occasionally, a disease <strong>of</strong> the blood cell system cannot be diagnosed and<br />

classified on the basis <strong>of</strong> the blood count alone and a bone marrow biopsy<br />

is indicated. In such cases it is more important to perform this biopsy competently<br />

and produce good smears for evaluation than to be able to interpret<br />

the bone marrow cytology yourself. Indications for bone marrow<br />

biopsy are given in Table 3.<br />

In the attempt to avoid complications, the traditional location for bone<br />

marrow biopsy at the sternum has been abandoned in favor <strong>of</strong> the superior<br />

part <strong>of</strong> the posterior iliac spine (back <strong>of</strong> the hipbone) (Fig. 5).<br />

Although the bone marrow cytology findings from the aspirate are sufficient<br />

or even preferable for most hematological questions (see Table 3), it<br />

is regarded as good practice to obtain a sample for bone marrow histology<br />

at the same time, since with improved instruments the procedure<br />

has become less stressful, and complementary cytological and histological<br />

data are then available from the start. After deep local anesthesia <strong>of</strong> the<br />

dorsal spine and a small skin incision, a histology cylinder at least 1.5 cm<br />

long is obtained using a sharp hollow needle (Yamshidi). A Klima and Rossegger<br />

cytology needle (Fig. 5) is then placed through the same subcutaneous<br />

channel but at a slightly different site from the earlier insertion<br />

point on the spine and gently pushed through the compacta. The mandrel<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Procedures, Assays, and Normal Values<br />

is pulled out and a 5- to 10-ml syringe body with 0.5 ml citrate or EDTA<br />

(heparin is used only for cytogenetics) is attached to the needle. The<br />

patient should be warned that there will be a painful drawing sensation<br />

during aspiration, which cannot be avoided. The barrel is then slowly<br />

pulled, and if the procedure is successful, blood from the bone marrow fills<br />

the syringe. The syringe body is separated from the needle and the mandrel<br />

reintroduced. The bone marrow aspirate is transferred from the syringe<br />

to a Petri dish. When the dish is gently shaken, small, pinhead-sized<br />

bone marrow spicules will be seen lying on the bottom. A smear, similar to<br />

a blood smear, can be prepared on a slide directly from the remaining contents<br />

<strong>of</strong> the syringe. If the first aspirate has obtained material, the needle is<br />

removed and a light compression bandage is applied.<br />

If the aspirate for cytology contains no bone marrow fragments (“punctio<br />

sicca,” dry tap), an attempt may be made to obtain a cytology smear<br />

from the (as yet unfixed) histology cylinder by rolling it carefully on the<br />

slide, but this seldom produces optimal results.<br />

The preparation <strong>of</strong> the precious bone marrow material demands<br />

special care. One or two bone marrow spicules are pushed to the outer<br />

edge <strong>of</strong> the Petri dish, using the mandrel from the sternal needle, a needle,<br />

or a wooden rod with a beveled tip, and transferred to a fat-free microscopy<br />

slide, on which they are gently pushed to and fro by the needle along<br />

the length <strong>of</strong> the slide in a meandering line. This helps the analyzing<br />

Fig. 5 Bone marrow biopsy from the superior part <strong>of</strong> the posterior iliac spine<br />

(back <strong>of</strong> the hipbone)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

21


22<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

Fig. 6 Squash preparation and meandering smear for the cytological analysis <strong>of</strong><br />

bone marrow spicules<br />

technician to make a differentiatial count. It should be noted that too<br />

much blood in the bone marrow sample will impede the semiquantitative<br />

analysis. In addition to this type <strong>of</strong> smear, squash preparations should also<br />

be prepared from the bone marrow material for selective staining. To do<br />

this, a few small pieces <strong>of</strong> bone marrow are placed on a slide and covered<br />

by a second slide. The two slides are lightly pressed and slid against each<br />

other, then separated (see Fig. 6).<br />

The smears are allowed to air-dry and some are incubated with panoptic<br />

Pappenheim staining solution (see previous text). Smears being sent to<br />

a diagnostic laboratory (wrapped individually and shipped as fragile<br />

goods) are better left unstained. Fresh smears <strong>of</strong> peripheral blood should<br />

accompany the shipment <strong>of</strong> each set <strong>of</strong> samples. (For principles <strong>of</strong> analysis<br />

and normal values see p. 52 ff., for indications for bone marrow cytology<br />

and histology see p. 27 ff.)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Procedures, Assays, and Normal Values<br />

Lymph Node Biopsy and Tumor Biopsy<br />

23<br />

These procedures, less invasive than bone marrow biopsy, are a simple<br />

and <strong>of</strong>ten diagnostically sufficient method for lymph node enlargement or<br />

other intumescences. The unanesthetized, disinfected skin is sterilized<br />

and pulled taut over the node. A no. 1 needle on a syringe with good suction<br />

is pushed through the skin into the lymph node tissue (Fig. 7). Tissue<br />

is aspirated from several locations, changing the angle <strong>of</strong> the needle<br />

slightly after each collection, and suction maintained while the needle is<br />

withdrawn into the subcutis. Aspiration ceases and the syringe is removed<br />

without suction. The biopsy harvest, which is in the needle, is extruded<br />

onto a microscopy slide and smeared out without force or pressure using a<br />

cover glass (spreader slide). Staining is done as described previously for<br />

blood smears.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


24<br />

1.<br />

Skin puncture<br />

2.<br />

Aspiration<br />

3.<br />

Collecting<br />

aspirates from<br />

different lymph<br />

node locations<br />

4.<br />

Detaching the<br />

syringe body,<br />

equalizing the<br />

pressure difference<br />

5.<br />

Removal <strong>of</strong> the<br />

syringe body and<br />

cannula<br />

6.<br />

Pulling back the<br />

syringe barrel<br />

7.<br />

Pushing the<br />

biopsy material<br />

onto a slide<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

Fig. 7 Procedure for<br />

lymph node biopsy<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Step-by-Step Diagnostic Sequence<br />

Step-by-Step Diagnostic Sequence<br />

On the basis <strong>of</strong> what has been said so far, the following guidelines for the<br />

diagnostic workup <strong>of</strong> hematological changes may be formulated:<br />

25<br />

1. The first step is quantitative determination <strong>of</strong> leukocytes (L), erythrocytes<br />

(E) and thrombocytes (T). Because the normal range can vary so widely<br />

in individual cases (Table 2), the following rule <strong>of</strong> thumb should be observed:<br />

A complete blood count (CBC) should be included in the baseline data,<br />

like blood pressure.<br />

2. All quantitative changes in L + E + T call for a careful evaluation <strong>of</strong> the<br />

differential blood count (DBC). Since clinical findings determine<br />

whether a DBC is indicated, it may be said that:<br />

A differential blood count is indicated:<br />

➤ By all unexplained clinical symptoms, especially<br />

➤ Enlarged lymph nodes or splenomegaly<br />

➤ Significant changes in any <strong>of</strong><br />

– Hemoglobin content or number <strong>of</strong> erythrocytes<br />

– Leukocyte count<br />

– Thrombocyte count<br />

The only initial assumption here is that mononuclear cells with unsegmented<br />

nuclei can be distinguished from polynuclear cells. While this<br />

nomenclature may not conform to ideal standards, it is well established<br />

and, moreover, <strong>of</strong> such practical importance as a fundamental distinguishing<br />

criterion that it is worth retaining. A marked majority <strong>of</strong><br />

mononuclear cells over the polynuclear segmented granulocytes is an<br />

unambiguous early finding.<br />

The next step has to be taken with far more care and discernment:<br />

classifying the mononuclear cells according to their possible origin, lymphatic<br />

cells, monocytes, or various immature blastic elements, which<br />

otherwise only occur in bone marrow. The aim <strong>of</strong> the images in the<br />

<strong>Atlas</strong> section <strong>of</strong> this book is to facilitate this part <strong>of</strong> the differential diagnosis.<br />

To a great extent, the possible origins <strong>of</strong> mononuclear cells can be<br />

distinguished; however, the limits <strong>of</strong> morphology and the vulnerability<br />

to artifacts are also apparent, leaving the door wide open to further<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


26<br />

Physiology and Pathophysiology <strong>of</strong> Blood Cells<br />

diagnostic steps (specialist morphological studies, cytochemistry,<br />

immunocytochemistry. A predominance <strong>of</strong> mononuclear cell elements<br />

has the same critical significance in the differential diagnosis <strong>of</strong> both<br />

leukocytoses and leukopenias.<br />

3. After the evaluation <strong>of</strong> the leukocytes, assessment <strong>of</strong> erythrocyte morphology<br />

is a necessary part <strong>of</strong> every blood smear evaluation. It is, naturally,<br />

particularly important in cases showing disturbances in the<br />

erythrocyte count or the hemoglobin.<br />

4. After careful consideration <strong>of</strong> the results obtained so far and the<br />

patient's clinical record, the last step is the analysis <strong>of</strong> the cell composition<br />

<strong>of</strong> the bone marrow. Quite <strong>of</strong>ten, suspected diagnoses are confirmed<br />

through humoral tests such as electrophoresis, or through cytochemical<br />

tests such as alkaline phosphatase, myeloperoxidase, nonspecific<br />

esterase, esterase, or iron tests.<br />

Bone marrow analysis is indicated when clinical findings and blood<br />

analysis leave doubts in the diagnostic assessment, for example in<br />

cases <strong>of</strong>:<br />

➤ Leukocytopenia<br />

➤ Thrombocytopenia<br />

➤ Undefined anemia<br />

➤ Tricytopenia, or<br />

➤ Monoclonal hypergammaglobulinemia<br />

A bone marrow analysis may be indicated in order to evaluate the<br />

spreading <strong>of</strong> a lymphadenoma or tumor, unless the bloodstream already<br />

shows the presence <strong>of</strong> pathological cells.<br />

5. Bone marrow histology is also rarely indicated (even more rarely than<br />

the bone marrow cytology). Examples <strong>of</strong> the decision-making process<br />

between bone marrow cytology and histology (biopsy) are shown in<br />

Table 3.<br />

Often only histological analysis can show structural changes or focal infiltration<br />

<strong>of</strong> the bone marrow.<br />

This is particularly true <strong>of</strong> the frequently fiber-rich chronic myeloproliferative<br />

diseases, such as polycythemia vera rubra, myel<strong>of</strong>ibrosis–<br />

osteomyelosclerosis (MF-OMS), essential thrombocythemia (ET), and<br />

chronic myeloid leukemia (CML) as well as malignant lymphoma<br />

without hematological involvement (Hodgkin disease or blastic non-<br />

Hodgkin lymphoma) and tumor infiltration.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Table 3 Indications for a differential blood count (DBC), bone marrow aspiration, and biopsy<br />

Indications Procedures<br />

All clinically unclear situations:<br />

➤ Enlarged lymph nodes or spleen<br />

➤ Changes in the simple CBC (penias or<br />

cytoses)<br />

➤ When a diagnosis cannot be made based on<br />

clinical findings and analysis <strong>of</strong> peripheral<br />

blood differential blood analysis, cytochemistry,<br />

phenotyping, molecular genetics or<br />

FISH<br />

Step-by-Step Diagnostic Sequence<br />

Differential blood count (DBC)<br />

Bone marrow analysis<br />

Bone marrow aspirate<br />

(Morphology, phenotyping,<br />

cytogenetics, FISH,<br />

molecular genetics)<br />

Bone marrow<br />

trephine biopsy<br />

(Morphology, immunohistology)<br />

➤ Punctio sicca ("dry tap") not possible +<br />

➤ Aplastic bone marrow not possible +<br />

➤ Suspicion <strong>of</strong> myelodysplastic syndrome + (+) (e. g. hypoplasia)<br />

➤ Pancytopenia + +<br />

➤ Anemia, isolated + –<br />

➤ Granulocytopenia, isolated + –<br />

➤ Thrombocytopenia, isolated (except ITP) + –<br />

➤ Suspected ITP (+) For therapy failure –<br />

➤ Suspected OMF/OMS – (BCR-ABL in peripheral<br />

blood is sufficient)<br />

+<br />

➤ Suspected PV – (BCR-ABL in peripheral<br />

blood is sufficient)<br />

–<br />

➤ Suspected ET – (BCR-ABL in peripheral<br />

blood is sufficient)<br />

+<br />

➤ Suspected CML – (BCR-ABL in peripheral (+) Conditions for the<br />

blood is sufficient) analysis<br />

➤ CMPE (BCR-ABL negative) (+) Differential diagnoses +<br />

➤ Suspected AL/AL + (+) Not in typical cases<br />

➤ Suspected bone marrow metastases – +<br />

➤ Monoclonal hypergammaglobulinemia + +<br />

➤ NHL (exceptions, see below) + +<br />

➤ Typical CLL + (+) Prognostic factor<br />

➤ Follicular lymphoma (+) To distinguish vs<br />

other NHL<br />

+<br />

➤ Hodgkin disease – +<br />

Further indications for a bone marrow analysis are:<br />

➤ Unexplained hypercalcemia + +<br />

➤ Inexplicable increase <strong>of</strong> bone AP – +<br />

➤ Obvious, unexplained abnormalities – +<br />

➤ Hyperparathyroidism – +<br />

➤ Paget disease – +<br />

➤ Osteomalacia – +<br />

➤ Renal osteopathy – +<br />

➤ Gaucher syndrome – +<br />

+ Recommended, – not recommended, (+) conditionally recommended, ITP idiopathic thrombocytopenia,<br />

OMF/OMS osteomyel<strong>of</strong>ibrosis/osteomyelosclerosis, PV polycythemia vera, ET essential thrombocythemia,<br />

CMPD chronic myeloproliferative disorders, AL acute leukemia, NHL non-Hodgkin lymphoma, CLL chronic<br />

lymphocytic leukemia, FISH fluorescence in situ hybridization, AP alkaline phosphatase.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

27


28<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Normal Cells <strong>of</strong> the Blood and<br />

Hematopoietic Organs<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


30<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

The Individual Cells <strong>of</strong> Hematopoiesis<br />

Immature Red Cell Precursors: Proerythroblasts and<br />

Basophilic Erythroblasts<br />

Proerythroblasts are the earliest, least mature cells in the erythrocyteforming<br />

series (erythropoiesis). Proerythroblasts are characterized by<br />

their size (about 20 µm), and by having a very dense nuclear structure<br />

with a narrow layer <strong>of</strong> cytoplasm, homogeneous in appearance, with a<br />

lighter zone at the center; they stain deep blue after Romanowsky staining.<br />

These attributes allow proerythroblasts to be distinguished from<br />

myeloblasts (p. 35) and thus to be assigned to the erythrocyte series. After<br />

mitosis, their daughter cells display similar characteristics except that<br />

they have smaller nuclei. Daughter cells are called basophilic erythroblasts<br />

(formerly also called macroblasts). Their nuclei are smaller and the chromatin<br />

is more coarsely structured.<br />

The maturation <strong>of</strong> cells in the erythrocyte series is closely linked to the<br />

activity <strong>of</strong> macrophages (transformed monocytes), which phagocytose<br />

nuclei expelled from normoblasts and iron from senescent erythrocytes,<br />

and pass these cell components on to developing erythrocytes.<br />

Diagnostic Implications. Proerythrocytes exist in circulating blood only<br />

under pathological conditions (extramedullary hematopoiesis; breakdown<br />

<strong>of</strong> the blood–bone marrow barrier by tumor metastases, p. 150; or<br />

erythroleukemia, p. 100). In these situations, basophilic erythroblasts may<br />

also occur; only exceptionally in the course <strong>of</strong> a strong postanemia regeneration<br />

will a very few <strong>of</strong> these be released into the blood stream (e.g.,<br />

in the compensation phase after severe hemorrhage or as a response to<br />

vitamin deficiency, see p. 152).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Normally erythropoiesis takes place only in the bone marrow<br />

a b<br />

Fig. 8 Early erythropoiesis. a The earliest recognizable red cell precursor is the<br />

large dark proerythroblast with loosely arranged nuclear chromatin (1). Below are<br />

two orthochromatic erythroblasts (2), on the right a metamyelocyte (3). b Proerythroblast<br />

(1). c Proerythroblast (1) next to a myeloblast (2) (see p. 34); lower<br />

region <strong>of</strong> image shows a promyelocyte (3). Toward the upper left are a metamyelocyte<br />

(4) and a segmented neutrophilic granulocyte (5).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

31<br />

c


32<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Mature Red Blood Precursor Cells: Polychromatic<br />

and Orthochromatic Erythroblasts (Normoblasts)<br />

and Reticulocytes<br />

The results <strong>of</strong> mitosis <strong>of</strong> erythroblasts are called normoblasts. This name<br />

covers two cell types with relatively dense round nuclei and grayish pink<br />

stained cytoplasm. The immature cells in which the cytoplasm displays a<br />

grayish blue hue, which are still able to divide, are now called “polychromatic<br />

erythroblasts,” while the cells in which the cytoplasm is already<br />

taking on a pink hue, which contain a lot <strong>of</strong> hemoglobin and are no longer<br />

able to divide, are called “orthochromatic erythroblasts.” The nuclei <strong>of</strong> the<br />

latter gradually condense into small black spheres without structural definition<br />

that eventually are expelled from the cells. The now enucleated<br />

young erythrocytes contain copious ribosomes that precipitate into reticular<br />

(“net-like”) structures after special staining (see p. 11), hence their<br />

name, reticulocytes.<br />

To avoid confusing erythroblasts and lymphoblasts (Fig. 9 d), note the<br />

completely rounded, very dense normoblast nuclei and homogeneous,<br />

unstructured cytoplasm <strong>of</strong> the erythroblasts.<br />

Diagnostic Implications. Polychromatic and orthochromatic erythroblasts<br />

may be released into the bloodstream whenever hematopoiesis is activated,<br />

e.g., in the compensation or treatment stage after hemorrhage or<br />

iron or vitamin deficiency. They are always present when turnover <strong>of</strong><br />

blood cells is chronically increased (hemolysis). Once increased blood regeneration<br />

has been excluded, the presence <strong>of</strong> erythroblasts in the blood<br />

should prompt consideration <strong>of</strong> two other disorders: extramedullary production<br />

<strong>of</strong> blood cells in myeloproliferative diseases (p. 114), and bone<br />

marrow carcinosis with destruction <strong>of</strong> the blood–bone marrow barrier<br />

(p. 154). In the same situations, the reticulocyte counts (after special staining)<br />

are elevated above the average <strong>of</strong> 25‰ for men and 40‰ for women,<br />

respectively, and can reach extremes <strong>of</strong> several hundred per mill.<br />

Fig. 9 Nucleated erythrocyte precursors. a Two basophilic erythroblasts with<br />

condensed chromatin structure (1) and a polychromatic erythroblast with an almost<br />

homogeneous nucleus (2). b The erythropoiesis in the bone marrow is <strong>of</strong>ten<br />

organized around a macrophage with a very wide, light cytoplasmic layer (1).<br />

Grouped around it are polychromatic erythroblasts <strong>of</strong> variable size. Erythroblast<br />

mitosis (2). c Polychromatic erythroblast (1) and orthochromatic erythroblast<br />

(normoblast) (2).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a b<br />

c<br />

d<br />

During increased turnover, nucleated red cell precursors may<br />

migrate into the peripheral blood<br />

Fig. 9 d The density <strong>of</strong> the nuclear chromatin is similar in lymphocytes (1) and<br />

erythroblasts (2), but in the erythroblast the cytoplasm is wider and similar in color<br />

to a polychromatic erythrocyte (3). e Normal red blood cell findings with slight<br />

variance in size <strong>of</strong> the erythrocytes. A lymphocyte (1) and a few thrombocytes (2)<br />

are seen. The erythrocytes are slightly smaller than the nucleus <strong>of</strong> the lymphocyte<br />

nucleus.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

33<br />

e


34<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Immature White Cell Precursors: Myeloblasts and<br />

Promyelocytes<br />

Myeloblasts are the least mature cells in the granulocyte lineage. Mononuclear,<br />

round-to-ovoid cells, they may be distinguished from proerythroblasts<br />

by the finer, “grainy” reticular structure <strong>of</strong> their nuclei and the<br />

faintly basophilic cytoplasm. On first impression, they may look like large<br />

or even small lymphocytes (micromyeloblasts), but the delicate structure<br />

<strong>of</strong> their nuclei always gives them away as myeloblasts. In some areas, condensed<br />

chromatin may start to look like nucleoli. Sporadically, the cytoplasm<br />

contains azurophilic granules.<br />

Promyelocytes are the product <strong>of</strong> myeloblast division, and usually grow<br />

larger than their progenitor cells. During maturation, their nuclei show an<br />

increasingly coarse chromatin structure. The nucleus is eccentric; the<br />

lighter zone over its bay-like indentation corresponds to the Golgi apparatus.<br />

The wide layer <strong>of</strong> basophilic cytoplasm contains copious large<br />

azurophilic granules containing peroxidases, hydrolases, and other<br />

enzymes. These granulations also exist scattered all around the nucleus, as<br />

may be seen by focusing on different planes <strong>of</strong> the preparation using the<br />

micrometer adjustment on the microscope.<br />

Diagnostic Implications. Ordinarily, both cell types are encountered only<br />

in the bone marrow, where they are the most actively dividing cells and<br />

main progenitors <strong>of</strong> granulocytes. In times <strong>of</strong> increased granulocyte production,<br />

promyelocytes and (in rare cases) myeloblasts may be released<br />

into the blood stream (pathological left shift, see p. 112). Under strong regeneration<br />

pressure from the erythrocyte series, too—e.g., during the<br />

compensation phase following various anemias—immature white cell<br />

precursors, like the red cell precursors, may be swept into the peripheral<br />

blood. Bone marrow involvement by tumor metastases also increases the<br />

permeability <strong>of</strong> the blood–bone marrow barrier for immature white cell<br />

precursors (for an overview, see p. 112 ff.).<br />

In some acute forms <strong>of</strong> leukemia, myeloblasts (and also, rarely, promyelocytes)<br />

dominate the blood analysis (p. 97).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Round cells with “grainy” reticular chromatin structure are<br />

blasts, not lymphocytes<br />

a b<br />

c d<br />

Fig. 10 Granulocyte precursors. a The least mature precursor in granulopoiesis<br />

is the myeloblast, which is released into the blood stream only under pathological<br />

conditions. A large myeloblast is shown with a fine reticular nuclear structure and<br />

a narrow layer <strong>of</strong> slightly basophilic cytoplasm without granules. b Myeloblast and<br />

neutrophilic granulocytes with segmented nuclei (blood smear from a patient<br />

with AML). c Myeloblast (1), which shows the start <strong>of</strong> azurophilic granulation<br />

(arrow), and a promyelocyte (2) with copious large azurophilic granules, typically<br />

in a perinuclear location. d Large promyelocyte (1), myelocyte (2), metamyelocyte<br />

(3), and polychromatic erythroblast (4).<br />

35<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


36<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Partly Mature White Cell Precursors: Myelocytes and<br />

Metamyelocytes<br />

Myelocytes are the direct product <strong>of</strong> promyelocyte mitosis and are always<br />

clearly smaller than their progenitors. The ovoid nuclei have a banded<br />

structure; the cytoplasm is becoming lighter with maturation and in some<br />

cases acquiring a pink tinge. A special type <strong>of</strong> granules, which no longer<br />

stain red like the granules in promyelocytes (“specific granules,” peroxidase-negative),<br />

are evenly distributed in the cytoplasm. Myelocyte morphology<br />

is wide-ranging because myelocytes actually cover three different<br />

varieties <strong>of</strong> dividing cells.<br />

Metamyelocytes (young granulocytes) are the product <strong>of</strong> the final myelocyte<br />

division and show further maturation <strong>of</strong> the nucleus with an increasing<br />

number <strong>of</strong> stripes and points <strong>of</strong> density that give the nuclei a spotted<br />

appearance. The nuclei slowly take on a kidney bean shape and have some<br />

plasticity. Metamyelocytes are unable to divide. From this stage on, only<br />

further maturation <strong>of</strong> the nucleus occurs by contraction, so that the distinctions<br />

(between metamyelocytes, band neutrophils, and segmented<br />

neutrophils) are merely conventional, although they do relate to the varying<br />

“maturation” <strong>of</strong> these cell forms.<br />

Diagnostic Implications. Like their precursors, myelocytes and metamyelocytes<br />

normally appear in the peripheral blood only during increased<br />

cell production in response to stress or triggers, especially infections (for<br />

an overview <strong>of</strong> possible triggers, see p. 112). Under these conditions, they<br />

are, however, more abundant than myeloblasts or promyelocytes.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a b<br />

c<br />

Myelocytes and metamyelocytes also occur in the blood stream<br />

in severe reactive disease<br />

Fig 11 Myelocytes and metamyelocytes. a Early myelocyte. The chromatin<br />

structure is denser than that <strong>of</strong> promyelocytes. The granules do not lie over the<br />

nucleus (as can be seen by turning the fine focus adjustment <strong>of</strong> the microscope to<br />

and fro). The blood smear is from a case <strong>of</strong> sepsis, hence the intensive granulation.<br />

b Slightly activated myelocyte (the cytoplasm is still relatively basophilic). c Typical<br />

myelocyte (1) close to a segmented neutrophil (2). d This metamyelocyte is<br />

distinguished from a myelocyte by incipient lobe formation.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

37<br />

d


38<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Mature Neutrophils: Band Cells and Segmented<br />

Neutrophils<br />

Band cells (band neutrophils) represent the further development <strong>of</strong><br />

metamyelocytes. Distinguishing between the different cell types is <strong>of</strong>ten<br />

difficult. The term “band cell” should be used when all nuclear sections <strong>of</strong><br />

the nucleus are approximately the same width (the “bands”). The beginnings<br />

<strong>of</strong> segmentation may be visible, but the indentations should never<br />

cut more than two-thirds <strong>of</strong> the way across the nucleus.<br />

Segmented neutrophils represent the final stage in the lineage that started<br />

with myeloblasts, forming gradually, without any clear transition or<br />

further cell divisions, by increasing contraction <strong>of</strong> their nuclei. Finally, the<br />

nuclear segments are connected only by narrow chromatin bridges, which<br />

should be no thicker than one-third <strong>of</strong> the average diameter <strong>of</strong> the nucleus.<br />

The chromatin in each segment forms coarse bands, or patches and<br />

is denser than the chromatin in band neutrophils.<br />

The cytoplasm <strong>of</strong> segmented neutrophilic granulocytes varies after<br />

staining from nearly colorless to s<strong>of</strong>t pink or violet. The abundant granules<br />

are <strong>of</strong>ten barely visible dots.<br />

The number <strong>of</strong> segments increases with the age <strong>of</strong> the cells. The following<br />

approximate values are taken to represent a normal distribution:<br />

10–30% have two segments, 40–50% have three segments, 10–20% have<br />

four segments, and 0–5% <strong>of</strong> the nuclei have five segments. A left shift to<br />

smaller numbers <strong>of</strong> segments is a discreet symptom <strong>of</strong> reactive activation<br />

<strong>of</strong> this cell series. A right shift to higher numbers <strong>of</strong> segments (oversegmentation)<br />

usually accompanies vitamin B12 and folic acid deficiencies.<br />

Diagnostic Implications. Banded neutrophilic granulocytes (band neutrophils)<br />

may occur in small numbers (up to 2%) in a normal blood count. This<br />

is <strong>of</strong> no diagnostic significance. A higher proportion than 2% may indicate<br />

a left shift and constitute the first sign <strong>of</strong> a reactive condition (p. 113). The<br />

diagnostic value <strong>of</strong> segmented neutrophilic granulocytes (segmented<br />

neutrophils) is that normal values are the most sensitive diagnostic indicator<br />

<strong>of</strong> normally functioning hematopoiesis (and, especially, <strong>of</strong> normal<br />

cellular defense against bacteria). An increase in segmented neutrophils<br />

without a qualitative left shift is not evidence <strong>of</strong> an alteration in bone marrow<br />

function, because under certain conditions stored cells may be released<br />

into the peripheral blood (for causes, see p. 111). In conjunction<br />

with qualitative changes (left shift, toxic granulations), however,<br />

granulocytosis does in fact indicate bone marrow activation that may have<br />

a variety <strong>of</strong> triggers (pp. 110 f.), and if the absolute number has fallen<br />

below the lower limit <strong>of</strong> the normal range (Table 2, p. 12), a bone marrow<br />

defect or increased cell death must be considered.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a b<br />

c<br />

Advancing nuclear contraction and segmentation: continuous<br />

transformation from metamyelocyte to band cell and then segmented<br />

neutrophilic granulocyte<br />

e<br />

Fig. 12 Neutrophils (neutrophilic granulocytes). a Transitional form between a<br />

metamyelocyte and a band cell. b Copious granulation in a band cell (1) (toxic granulation)<br />

next to band cells (2) with Döhle bodies (arrows). c Two band cells.<br />

d Band cells can also occur as aggregates. e Segmented neutrophilic granulocytes.<br />

f Segmented neutrophilic granulocyte after the peroxidase reaction.<br />

g Segmented neutrophilic granulocyte after alkaline leukocyte phosphatase (ALP)<br />

staining.<br />

39<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

d<br />

f<br />

g


40<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Cell Degradation, Special Granulations, and Nuclear<br />

Appendages in Neutrophilic Granulocytes and Nuclear<br />

Anomalies<br />

Toxic granulation is the term used when the normally faint stippled<br />

granules in segmented neutrophils stain an intense reddish violet, usually<br />

against a background <strong>of</strong> slightly basophilic cytoplasm; unlike the normal<br />

granules, they stain particularly well in an acidic pH (5.4). This phenomenon<br />

is a consequence <strong>of</strong> activity against bacteria or proteins and is observed<br />

in serious infections, toxic or drug effects, or autoimmune<br />

processes (e.g., chronic polyarthritis). At the same time, cytoplasmic<br />

vacuoles are <strong>of</strong>ten found, representing the end stage <strong>of</strong> phagocytosis (especially<br />

in cases <strong>of</strong> sepsis), as are Döhle bodies: small round bodies <strong>of</strong> basophilic<br />

cytoplasm that have been described particularly in scarlet fever,<br />

but may be present in all serious infections and toxic conditions. A deficiency<br />

or complete absence <strong>of</strong> granulation in neutrophils is a sign <strong>of</strong><br />

severe disturbance <strong>of</strong> the maturation process (e.g., in myelodysplasia or<br />

acute leukemia). The Pelger anomaly, named after its first describer, is a<br />

hereditary segmentation anomaly <strong>of</strong> granulocytes that results in round,<br />

rod-shaped, or bisegmented nuclei. The same appearance as a nonhereditary<br />

condition (pseudo-Pelger formation, also called Pel–Ebstein fever, or<br />

[cyclic] Murchison syndrome) indicates a severe infectious or toxic stress<br />

response or incipient myelodysplasia; it also may accompany manifest<br />

leukemia.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Note the granulations, inclusions, and appendages in segmented<br />

neutrophilic granulocytes<br />

a b<br />

c d<br />

Fig. 13 Variations <strong>of</strong> segmented neutrophilic granulocytes. a Reactive state with<br />

toxic granulation <strong>of</strong> the neutrophilic granulocytes, more visibly expressed in the<br />

cell on the left (1) than the cell on the right (2) (compare with nonactivated cells,<br />

p. 39). b Sepsis with toxic granulation, cytoplasmic vacuoles, and Döhle bodies<br />

(arrows) in band cells (1) and a monocyte (2). c Pseudo-Pelger cell looking like<br />

sunglasses (toxic or myelodysplastic cause). d Döhle-like basophilic inclusion (arrow)<br />

without toxic granulation. Together with giant thrombocytes this suggests<br />

May–Hegglin anomaly. continued <br />

41<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


42<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Nuclear appendages, which must not to be mistaken for small segments,<br />

are minute (less than the size <strong>of</strong> a thrombocyte) chromatin bodies that remain<br />

connected to the main part <strong>of</strong> the nucleus via a thin bridge and consequently<br />

look like a drumstick, sessile nodule, or small tennis racket. Of<br />

these, only the drumstick form corresponds to the X-chromosome, which<br />

has become sequestered during the process <strong>of</strong> segmentation. A proportion<br />

<strong>of</strong> 1–5% circulating granulocytes with drumsticks (at least 6 out <strong>of</strong> 500)<br />

suggests female gender; however, because the drumstick form is easy to<br />

confuse with the other (insignificant) forms <strong>of</strong> nuclear appendage, care<br />

should be taken before jumping to conclusions.<br />

Rarely, degrading forms <strong>of</strong> granulocytes, shortly before cytolysis or<br />

apoptosis, may be found in the blood (they are more frequent in exudates).<br />

In these, the segments <strong>of</strong> the nucleus are clearly losing connection, and the<br />

chromatin structure <strong>of</strong> the individual segments, which are becoming<br />

round, becomes dense and homogeneous.<br />

Diagnostic Implications. Toxic granulation indicates bacterial, chemical, or<br />

metabolic stress. Pseudo-Pelger granulocytes are observed in cases <strong>of</strong> infectious–toxic<br />

stress conditions, myelodysplasia, and leukemia.<br />

The use <strong>of</strong> nuclear appendages to determine gender has lost significance<br />

in favor <strong>of</strong> genetic testing.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Note the granulations, inclusions, and appendages in segmented<br />

neutrophilic granulocytes<br />

e f<br />

g h<br />

Fig. 13 continued. e Hypersegmented neutrophilic granulocyte (six or more<br />

segments). There is an accumulation <strong>of</strong> these cells in megaloblastic anemia. f<br />

Drumstick (arrow 1) as an appendage with a thin filament bridge to the nucleus<br />

(associated with the X-chromosome), adjoined by a thrombocyte (arrow 2). g<br />

Very large granulocyte from a blood sample taken after chemotherapy. h Segmented<br />

neutrophilic granulocyte during degradation, <strong>of</strong>ten seen as an artifact after<br />

prolonged sample storage (more than eight hours).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

43


44<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Eosinophilic Granulocytes (Eosinophils)<br />

Eosinophils arise from the same stem cell population as neutrophils and<br />

mature in parallel with them. The earliest point at which eosinophils can<br />

be morphologically defined in the bone marrow is at the promyelocyte<br />

stage. Promyelocytes contain large granules that stain blue–red; not until<br />

they reach the metamyelocyte stage do these become a dense population<br />

<strong>of</strong> increasingly round, golden-red granules filling the cytoplasm. The<br />

Charcot–Leyden crystals found between groups <strong>of</strong> eosinophils in exudates<br />

and secretions have the same chemical composition as the eosinophil<br />

granules.<br />

The nuclei <strong>of</strong> mature eosinophils usually have only two segments.<br />

Diagnostic Implications. In line with their function (see p. 5) (reaction<br />

against parasites and regulation <strong>of</strong> the immune response, especially<br />

defense against foreign proteins), an increase <strong>of</strong> eosinophils above 400/µl<br />

should be seen as indicating the presence <strong>of</strong> parasitosis, allergies, and<br />

many other conditions (p. 124).<br />

Basophilic Granulocytes (Basophils)<br />

Like eosinophils, basophils (basophilic granulocytes) mature in parallel<br />

with cells <strong>of</strong> the neutrophil lineage. The earliest stage at which they can be<br />

identified is the promyelocyte stage, at which large, black–violet stained<br />

granules are visible. In mature basophils, which are relatively small, these<br />

granules <strong>of</strong>ten overlie the two compact nuclear segments like blackberries.<br />

However, they easily dissolve in water, leaving behind faintly pink<br />

stained vacuoles.<br />

Close relations <strong>of</strong> basophilic granulocytes are tissue basophils or tissue<br />

mast cells—but these are never found in blood. Tissue basophils have a<br />

round nucleus underneath large basophilic granules.<br />

Diagnostic Implications. In line with their role in anaphylactic reactions<br />

(p. 5), elevated basophil counts are seen above all in hypersensitivity reactions<br />

<strong>of</strong> various kinds. Basophils are also increased in chronic myeloproliferative<br />

bone marrow diseases, especially chronic myeloid leukemia<br />

(pp. 117, 120).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

e<br />

Round granules filling the cytoplasm: eosinophilic and basophilic<br />

granulocytes<br />

Fig. 14 Eosinophilic and basophilic granulocytes. a–c Eosinophilic granulocytes<br />

with corpuscular, orange-stained granules. d In contrast, the granules <strong>of</strong> neutrophilic<br />

granulocytes are not round but more bud-shaped. e Basophilic granulocyte.<br />

The granules are corpuscular like those <strong>of</strong> the eosinophilic granulocyte but stain<br />

deep blue to violet. f Very prominent large granules in a basophilic granulocyte in<br />

chronic myeloproliferative disease.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

45<br />

b<br />

d<br />

f


46<br />

Monocytes<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Monocytes are produced in the bone marrow; their line <strong>of</strong> development<br />

branches <strong>of</strong>f at a very early stage from that <strong>of</strong> the granulocytic series (see<br />

flow chart p. 3), but does not contain any distinct, specific precursors that<br />

can be securely identified with diagnostic significance in everyday morphological<br />

studies. Owing to their great motility and adhesiveness, mature<br />

monocytes are morphologically probably the most diversified <strong>of</strong> all<br />

cells. Measuring anywhere between 20 and 40 µm in size, their constant<br />

characteristic is an ovoid nucleus, usually irregular in outline, with invaginations<br />

and <strong>of</strong>ten pseudopodia-like cytoplasmic processes. The fine,<br />

“busy” structure <strong>of</strong> their nuclear chromatin allows them to be distinguished<br />

from myelocytes, whose chromatin has a patchy, streaky structure,<br />

and also from lymphocytes, which have dense, homogeneous nuclei.<br />

The basophilic cytoplasmic layer varies in width, stains a grayish color,<br />

and contains a scattered population <strong>of</strong> very fine reddish granules that are<br />

at the very limit <strong>of</strong> the eye’s resolution. These characteristics vary greatly<br />

with the size <strong>of</strong> the monocyte, which in turn is dependent on the thickness<br />

<strong>of</strong> the smear. Where the smear is thin, especially at the feathered end,<br />

monocytes are abundant, relatively large and loosely structured, and their<br />

cytoplasm stains light gray–blue (“dove gray”). In thick, dense parts <strong>of</strong> the<br />

smear, some monocytes look more like lymphocytes: only a certain nuclear<br />

indentation and the “thundercloud” gray–blue staining <strong>of</strong> the cytoplasm<br />

may still mark them out.<br />

Diagnostic Implications. In line with their function (see p. 5) as phagocytic<br />

defense cells, an elevation <strong>of</strong> the monocyte population above 7% and<br />

above 850/µl indicates an immune defense reaction; only when a sharp<br />

rise in monocyte counts is accompanied by a drop in absolute counts in<br />

the other cell series is monocytic leukemia suggested (p. 101). Phagocytosis<br />

<strong>of</strong> erythrocytes and white blood cells (hemophagocytosis) may occur<br />

in some virus infections and autoimmune diseases.<br />

➤ Monocytosis in cases <strong>of</strong> infection: always present at the end <strong>of</strong> acute infections;<br />

chronic especially in<br />

– Endocarditis lenta, listeriosis, brucellosis, tuberculosis<br />

➤ Monocytosis in cases <strong>of</strong> a non-infectious response, e.g.,<br />

– Collagenosis, Crohn disease, ulcerative colitis<br />

➤ Monocytoses in/as neoplasia, e.g.,<br />

– Paraneoplastic in cases <strong>of</strong> disseminating tumors, bronchial carcinoma,<br />

breast carcinoma, Hodgkin disease, myelodysplasias (especially<br />

CMML, pp. 107 f) and acute monocytic leukemia (p. 101)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Monocytes show the greatest morphological variation among<br />

blood cells<br />

a b<br />

c d<br />

e f<br />

g h<br />

Fig. 15 Monocytes. a–c Range <strong>of</strong> appearances <strong>of</strong> typical monocytes with lobed,<br />

nucleus, gray–blue stained cytoplasm and fine granulation. d Phagocytic monocyte<br />

with plasma vacuoles. e Monocyte (1) to the right <strong>of</strong> a lymphocyte with azurophilic<br />

granules (2). f Monocyte (1) with nucleus resembling that <strong>of</strong> a band neutrophil,<br />

but its cytoplasm stains typically gray–blue. Lymphocyte (2). g A monocyte<br />

that has phagocytosed two erythrocytes and harbors them in its wide cytoplasm<br />

(arrows) (sample taken after bone marrow transplantation). h Esterase<br />

staining, a typical marker enzyme for cells <strong>of</strong> the monocyte lineage.<br />

47<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


48<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Lymphocytes (and Plasma Cells)<br />

Lymphocytes are produced everywhere, particularly in the lymph nodes,<br />

spleen, bone marrow, and the lymphatic islands <strong>of</strong> the intestinal mucosa,<br />

under the influence <strong>of</strong> the thymus (T-lymphocytes, about 80%), or the<br />

bone marrow (B-lymphocytes, about 20%). A small fraction <strong>of</strong> the lymphocytes<br />

are NK cells (natural killer cells). Immature precursor cells (lymph<br />

node cytology, p. 177) are practically never released into the blood and are<br />

therefore <strong>of</strong> no practical diagnostic significance. The cells encountered in<br />

circulating blood are mostly “small” lymphocytes with oval or round nuclei<br />

6–9 µm in diameter. Their chromatin may be described as dense and<br />

coarse. Detailed analysis under the microscope, using the micrometer<br />

screw to view the chromatin in different planes, reveals not the patch-like<br />

or banded structure <strong>of</strong> myeloblast chromatin, or the “busy” structure <strong>of</strong><br />

monocyte chromatin, but slate-like formations with homogeneous chromatin<br />

and intermittent narrow, lighter layers that resemble geological<br />

break lines. Nucleoli are rarely seen. The cytoplasm wraps quite closely<br />

around the nucleus and is slightly basophilic. Only a few lymphocytes display<br />

the violet stained stippling <strong>of</strong> granules; about 5% <strong>of</strong> small lymphocytes<br />

and about 3% <strong>of</strong> large ones. The family <strong>of</strong> large lymphocytes with<br />

granulation consists mostly <strong>of</strong> NK cells. An important point is that small<br />

lymphocytes—which cannot be identified as T- or B-lymphocytes on the<br />

basis <strong>of</strong> morphology—are not functional end forms, but undergo transformation<br />

in response to specific immunological stimuli. The final stage <strong>of</strong> Blymphocyte<br />

maturation (in bone marrow and lymph nodes) is plasma<br />

cells, whose nuclei <strong>of</strong>ten show radial bars, and whose basophilic cytoplasm<br />

layer is always wide. Intermediate forms (“plasmacytoid” lymphocytes)<br />

also exist.<br />

Diagnostic Implications. Values between 1500 and 4000/µl and about 35%<br />

reflect normal output <strong>of</strong> the lymphatic system. Elevated absolute lymphocyte<br />

counts, <strong>of</strong>ten along with cell transformation, are observed predominantly<br />

in viral infections (pp. 67, 69) or in diseases <strong>of</strong> the lymphatic system<br />

(p. 75 ff.). Relative increases at the expense <strong>of</strong> other blood cell series may<br />

be a manifestation <strong>of</strong> toxic or aplastic processes (agranulocytosis, p. 87;<br />

aplastic anemia, p. 148), because these irregularities are rare in the lymphatic<br />

series. A spontaneous decrease in lymphocyte counts is normally<br />

seen only in very rare congenital diseases (agammaglobulinemia [Bruton<br />

disease], DiGeorge disease [chromosome 22q11 deletion syndrome]).<br />

Some systemic diseases also lead to low lymphocyte counts (Hodgkin disease,<br />

active AIDS).<br />

Mature plasma cells are rarely found in blood (plasma cell leukemia is<br />

extremely rare). Plasma-cell-like (“plasmacytoid”) lymphocytes occur in<br />

viral infections or systemic diseases (see p. 68 f. and p. 74 f.).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

b c<br />

d<br />

Lymphocytes are small round cells with dense nuclei and some<br />

variation in their appearance<br />

f<br />

g<br />

Fig. 16 Lymphocytes a–c Range <strong>of</strong> appearance <strong>of</strong> normal lymphocytes (some <strong>of</strong><br />

them adjacent to segmented neutrophilic granulocytes). d In neonates, some<br />

lymphocytes from a neonate show irregularly shaped nuclei with notches or hints<br />

<strong>of</strong> segmentation. e A few larger lymphocytes with granules may occur in a normal<br />

person. f Occasionally, and without any recognizable trigger, the cytoplasm may<br />

widen. g A smear taken after infection may contain a few plasma cells, the final,<br />

morphologically fully developed cells in the B-lymphocyte series (for further activated<br />

lymphocyte forms, see p. 67).<br />

49<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

e


50<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Megakaryocytes and Thrombocytes<br />

Megakaryocytes can enter the bloodstream only in highly pathological<br />

myeloproliferative disease or acute leukemia. They are shown here in<br />

order to demonstrate thrombocyte differentiation. Megakaryocytes reside<br />

in the bone marrow and have giant, extremely hyperploid nuclei (16<br />

times the normal number <strong>of</strong> chromosome sets on average), which build up<br />

by endomitosis. Humoral factors regulate the increase <strong>of</strong> megakaryocytes<br />

and the release <strong>of</strong> thrombocytes when more are needed (e.g., bleeding or<br />

increased thrombocyte degradation). Cytoplasm with granules is pinched<br />

<strong>of</strong>f from megakaryocytes to form thrombocytes. The residual naked megakaryocyte<br />

nuclei are phagocytosed.<br />

Only mature thrombocytes occur in blood. About 1–4 µm in size and<br />

anuclear, their light blue stained cytoplasm and its processes give them a<br />

star-like appearance, with fine reddish blue granules near the center.<br />

Young thrombocytes are larger and more “spread out;” older ones look<br />

like pyknotic dots.<br />

Diagnostic Implications. In a blood smear, there are normally 8–15 thrombocytes<br />

per view field using a 100 objective; they may appear dispersed<br />

or in groups. To someone quickly screening a smear, they will give a good<br />

indication <strong>of</strong> any increase or strong decrease in the count, which can be<br />

useful for early diagnosis <strong>of</strong> acute thrombocytopenias (p. 164 f.).<br />

Small megakaryocyte nuclei are found in the bloodstream only in<br />

severe myeloproliferative disorders (p. 171).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Megakaryocytes are never present in a normal peripheral blood<br />

smear. Thrombocytes are seen in every field view<br />

a b<br />

c d<br />

Fig. 17 Megakaryocytes and thrombocytes. a Megakaryocytes in a bone marrow<br />

smear. The wide cytoplasm displays fine, cloudy granulation as a sign <strong>of</strong> incipient<br />

thrombocyte budding. b Normal density <strong>of</strong> thrombocytes among the erythrocytes,<br />

with little variation in thrombocyte size. c and d Peripheral blood smears<br />

with aggregations <strong>of</strong> thrombocytes. When such aggregates are seen against a<br />

background <strong>of</strong> apparent thrombocytopenia, the phenomenon is called “pseudothrombocytopenia”<br />

and is usually an effect <strong>of</strong> the anticoagulant EDTA (see also<br />

p. 167).<br />

51<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


52<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Bone Marrow: Cell Composition<br />

and Principles <strong>of</strong> Analysis<br />

As indicated above, and as will be shown below, almost all disorders <strong>of</strong> the<br />

hematopoietic system can be diagnosed using clinical findings, blood<br />

analysis, and humoral data. There is no mystery about bone marrow diagnostics.<br />

The basic categories are summarized here to give an understanding<br />

<strong>of</strong> how specific diagnostic information is achieved; photomicrographs<br />

will show the appearance <strong>of</strong> specific diseases. Once the individual cell<br />

types, as given in the preceding pages, are recognized, it becomes possible<br />

to interpret the bone marrow smears that accompany the various diseases,<br />

and to follow further, analogous diagnostic steps. As a first step in<br />

the analysis <strong>of</strong> a bone marrow tissue smear or squash preparation, various<br />

areas in several preparations are broadly surveyed. This is followed by individual<br />

analysis <strong>of</strong> at least 200 cells from two representative areas. Table<br />

4 shows the mean normal values and their wide ranges.<br />

A combination <strong>of</strong> estimation and quantitative analysis is used, based on<br />

the following criteria:<br />

Cell Density. This parameter is very susceptible to artifacts. Figure 18<br />

shows roughly the normal cell density. A lower count may be due to the<br />

manner in which the sample was obtained or to the smearing procedure. A<br />

bone marrow smear typically shows areas where connective tissue adipocytes<br />

with large vacuoles predominate. Only if these adipocytes areas are<br />

present is it safe to assume that the smear contains bone marrow material<br />

and that an apparent deficit <strong>of</strong> bone marrow cells is real.<br />

Increased cell density: e.g., in all strong regeneration or compensation<br />

processes, and in cases <strong>of</strong> leukemia and myeloproliferative syndromes<br />

(except osteomyelosclerosis).<br />

Decreased cell density: e.g., in aplastic processes and myel<strong>of</strong>ibrosis.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Bone Marrow: Cell Composition and Principles <strong>of</strong> Analysis<br />

Table 4 Cell composition in the bone marrow: normal values (%)<br />

Median<br />

values<br />

(J. Boll)<br />

Median values and normal<br />

range<br />

(K. Rohr)<br />

Red cell series<br />

– Proerythrocytes 1<br />

– Macroblasts (basophilic<br />

erythroblasts)<br />

3 3.5 0.5–7.5<br />

– Normoblasts (poly- and<br />

orthochromic erythroblasts)<br />

16 19 (7–40)<br />

Neutrophil series<br />

– Myeloblasts 2.7 1 (0.5–5)<br />

– Promyelocytes 9.5 3 (0–7.5)<br />

– Myelocytes 14 15 (5–25)<br />

– Metamyelocytes 10.5 15 (5–20)<br />

– Band neutrophils 9.8 15 (5–25)<br />

– Segmented neutrophils 17.5 7 (0.5–15)<br />

Small cell series<br />

– Eosinophilic granulocytes 5 3 (1–7)<br />

– Basophilic granulocytes 1 0.5 (0–1)<br />

– Monocytes 2 2 (0.5–3)<br />

– Lymphocytes 6 7.5 (2.5–15)<br />

– Plasma cells 1.5 1 (0.5–3)<br />

Megakaryocytes<br />

Cell densities vary widely, 0.5–2 per view field during screening at low magnification.<br />

53<br />

Ratios <strong>of</strong> Red Cell Series to White Cell Series. In the final analysis, bone<br />

marrow cytology allows a quantitative assessment only in relative terms.<br />

The important ratio <strong>of</strong> red precursor cells to white cells is 1 : 2 for men<br />

and 1 : 3 for women.<br />

Shifts towards erythropoiesis are seen in all regenerative anemias (hemorrhagic<br />

anemia, iron deficiency anemia, vitamin deficiency anemia, and<br />

hemolysis), pseudopolycythemia (Gaisböck syndrome), and polycythemia,<br />

also in rare pseudo-regenerative disorders, such as sideroachrestic<br />

anemia and myelodysplasias. Shifts toward granulopoiesis are seen<br />

in all reactive processes (infections, tumor defense) and in all malignant<br />

processes <strong>of</strong> the white cell series (chronic myeloid leukemia, acute<br />

leukoses).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


54<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Distribution and Cell Quality in Erythropoiesis. In erythropoiesis polychromatic<br />

erythroblasts normally predominate. Proerythroblasts and basophilic<br />

erythroblasts only make up a small portion (Table 4). Here, too, a<br />

left shift indicates an increase in immature cell types and a right shift an<br />

increase in orthochromatic erythroblasts. Qualitatively, vitamin B12 and<br />

folic acid deficiency lead to a typical loosening-up <strong>of</strong> the nuclear structure<br />

in proerythroblasts and to nuclear segmentation and break-up in the<br />

erythroblasts (megaloblastic erythropoiesis).<br />

A left shift is seen in regenerative anemias except hemolysis. Atypical<br />

proerythroblasts predominate in megaloblastic anemia and erythremia.<br />

A right shift is seen in hemolytic conditions (nests <strong>of</strong> normoblasts, erythrons).<br />

Distribution and Cell Quality in Granulopoiesis. The same principle is valid<br />

as for erythropoiesis: the more mature the cells, the greater proportion <strong>of</strong><br />

the series they make up. A left shift indicates a greater than normal proportion<br />

<strong>of</strong> immature cells and a right shift a greater than normal proportion<br />

<strong>of</strong> mature cells (Table 4). Strong reactive conditions may lead to dissociations<br />

in the maturation process, e.g., the nucleus shows the structure <strong>of</strong><br />

a myelocyte while the cytoplasm is still strongly basophilic. In malignancies,<br />

the picture is dominated by blasts, which may <strong>of</strong>ten be difficult to<br />

identify with any certainty.<br />

A left shift is observed in all reactive processes and at the start <strong>of</strong> neoplastic<br />

transformation (smoldering anemia, refractory anemia with<br />

excess blasts [RAEB]). In acute leukemias, undifferentiated and partially<br />

matured blasts may predominate. In agranulocytosis, promyelocytes are<br />

most abundant.<br />

A right shift is diagnostically irrelevant.<br />

Cytochemistry. To distinguish between reactive processes and chronic<br />

myeloid leukemia, leukocyte alkaline phosphatase is determined in fresh<br />

smears <strong>of</strong> blood. To distinguish between different types <strong>of</strong> acute leukemia,<br />

the peroxidase and esterase reactions are carried out (pp. 97 and 99), and<br />

iron staining is performed (p. 109) if myelodysplasia is suspected.<br />

Cytogenetic Analysis. This procedure will take the diagnosis forward in<br />

cases <strong>of</strong> leukemia and some lymphadenomas. The fresh material must be<br />

heparinized before shipment, preferably after discussion with a specialist<br />

laboratory.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a b<br />

c<br />

The bone marrow contains a mixture <strong>of</strong> all the hematopoietic<br />

cells<br />

Fig. 18 Bone marrow cytology. a Bone marrow cytology <strong>of</strong> normal cell density in<br />

a young adult (smear from a bone marrow spicule shown at the lower right; magnification<br />

100). b More adipocytes with large vacuoles are present in this bone<br />

marrow preparation with normal hematopoietic cell densities; usually found in<br />

older patients. c Normal bone marrow cytology (magnification 400). Even this<br />

overview shows clearly that erythropoiesis (dense, black, round nuclei) accounts<br />

for only about one-third <strong>of</strong> all the cells.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

55


56<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Qualitative and Quantitative Assessment <strong>of</strong> the Remaining Cells. Lymphocyte<br />

counts may be slightly raised in reactive processes, but a significant<br />

increase suggests a disease <strong>of</strong> the lymphatic system. The exact classification<br />

<strong>of</strong> these disease follows the criteria <strong>of</strong> lymphocyte morphology<br />

(Fig. 16). If elevated lymphocyte counts are found only in one preparation<br />

or within a circumscribed area, physiological lymph follicles in the bone<br />

marrow are likely to be the source. In a borderline case, the histology and<br />

analysis <strong>of</strong> lymphocyte surface markers yield more definitive data.<br />

Plasma cell counts are also slightly elevated in reactive processes and<br />

very elevated in plasmacytoma. Reactive increase <strong>of</strong> lymphocytes and<br />

plasma cells with concomitant low counts in the other series is <strong>of</strong>ten an<br />

indication <strong>of</strong> panmyelopathy (aplastic anemia).<br />

Raised eosinophil and monocyte counts in bone marrow have the same<br />

diagnostic significance as in blood (p. 44).<br />

Megakaryocyte counts are reduced under the effects <strong>of</strong> all toxic stimuli<br />

on bone marrow. Counts increase after bleeding, in essential thrombocytopenia<br />

(Werlh<strong>of</strong> syndrome), and in myeloproliferative diseases (chronic<br />

myeloid leukemia, polycythemia, and essential thrombocythemia).<br />

Iron Staining <strong>of</strong> Erythropoietic Cells. Perls’ Prussian blue (also known as<br />

Perls’ acid ferrocyanide reaction) shows the presence <strong>of</strong> ferritin in 20–40%<br />

<strong>of</strong> all normoblasts, in the form <strong>of</strong> one to four small granules. The ironcontaining<br />

cells are called sideroblasts. Greater numbers <strong>of</strong> ferritin<br />

granules in normoblasts indicate a disorder <strong>of</strong> iron utilization (sideroachresia,<br />

especially in myelodysplasia), particularly when the granules<br />

form a ring around the nucleus (ring sideroblasts). Perls’ Prussian blue reaction<br />

also stains the diffuse iron precipitates in macrophages (Fig. 19 b).<br />

Under exogenous iron deficiency conditions the proportion <strong>of</strong> sideroblasts<br />

and iron-storing macrophages is reduced. However, if the shift in<br />

iron utilization is due to infectious and/or toxic conditions, the iron content<br />

in normoblasts is reduced while the macrophages are loaded with<br />

iron to the point <strong>of</strong> saturation. In hemolytic conditions, the iron content <strong>of</strong><br />

normoblasts is normal; it is elevated only in essential or symptomatic refractory<br />

anemia (including megaloblastic anemia).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Not just the individual cell, but its relative proportion is relevant<br />

in bone marrow diagnostics<br />

a b<br />

c d<br />

Fig. 19 Normal bone marrow findings. a Normal bone marrow: megakaryocyte<br />

(1), erythroblasts (2), and myelocyte (3). b Iron staining in the bone marrow cytology:<br />

iron-storing macrophage. c Normal bone marrow with slight preponderance<br />

<strong>of</strong> granulocytopoiesis, e.g., promyelocyte (1), myelocyte (2), metamyelocyte (3),<br />

and band granulocyte (4). d Normal bone marrow with slight preponderance <strong>of</strong><br />

erythropoiesis, e.g., basophilic erythroblast (1), polychromatic erythroblasts (2),<br />

and orthochromatic erythroblast (3). Compare (differential diagnosis) with the<br />

plasma cell (4) with its eccentric nucleus.<br />

57<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


58<br />

Normal Cells <strong>of</strong> the Blood and Hematopoietic Organs<br />

Bone Marrow: Medullary Stroma Cells<br />

➤ Fibroblastic reticular cells form a firm but elastic matrix in which the<br />

blood-forming cells reside, and are therefore rarely found in the bone<br />

marrow aspirate or cytological smear. When present, they are most<br />

likely to appear as dense cell groups with long fiber-forming cytoplasmic<br />

processes and small nuclei. Iron staining shows them up as a<br />

group <strong>of</strong> reticular cells which, like macrophages, have the potential to<br />

store iron. If they become the prominent cell population in the bone<br />

marrow, an aplastic or toxic medullary disorder must be considered.<br />

➤ Reticular histiocytes (not yet active in phagocytosis) are identical to<br />

phagocytic macrophages and are the main storage cells for tissuebound<br />

iron. Because <strong>of</strong> their small nuclei and easy-flowing cytoplasm,<br />

they are noticeable after panoptic staining only when they contain obvious<br />

entities such as lipids or pigments.<br />

➤ Osteoblasts are large cells with wide, eccentric nuclei. They differ from<br />

plasma cells in that the cytoplasm has no perinuclear lighter space (cell<br />

center) and stains a cloudy grayish-blue. As they are normally rare in<br />

bone marrow, increased presence <strong>of</strong> osteoblasts in the marrow may indicate<br />

metastasizing tumor cells (from another location).<br />

➤ Osteoclasts are multinucleated syncytia with wide layers <strong>of</strong> grayishblue<br />

stained cytoplasm, which <strong>of</strong>ten displays delicate azurophilic<br />

granulation. They are normally extremely rare in aspirates, and when<br />

they are found it is usually under the same conditions as osteoblasts.<br />

They are distinguished from megakaryocytes by their round and regular<br />

nuclei and by their lack <strong>of</strong> thrombocyte buds.<br />

From the above, it will be seen that bone marrow findings can be assessed<br />

on the basis <strong>of</strong> a knowledge <strong>of</strong> the cells elements described above<br />

(p. 32 ff.), taking account <strong>of</strong> the eight categories given on p. 52 f. It also<br />

shows that a diagnosis from bone marrow aspirates can safely be made<br />

only in conjunction with clinical findings, blood chemistry, and the qualitative<br />

and quantitative blood values. For this reason, a table <strong>of</strong> diagnostic<br />

steps taking account <strong>of</strong> these categories is provided at the beginning <strong>of</strong><br />

each <strong>of</strong> the following chapters.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Cells from the bone marrow stroma never occur in the blood<br />

stream<br />

Fig. 20 Bone marrow stroma. a Spindle-shaped fibroblasts form the structural<br />

framework <strong>of</strong> the bone marrow (shown here: aplastic hematopoiesis after therapy<br />

for multiple myeloma). b A macrophage has phagocytosed residual nuclear<br />

material (here after chemotherapy for acute leukemia). c Bone marrow osteoblasts<br />

are rarely found in the cytological assessment. The features that distinguish<br />

osteoblasts from plasma cells are their more loosely structured nuclei and the<br />

cloudy, “busy” basophilic cytoplasm. d Osteoclasts are multinucleated giant cells<br />

with wide, spreading cytoplasm.<br />

59<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

b<br />

d


60<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Abnormalities <strong>of</strong> the White Cell Series<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


62<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

A very old-fashioned, intuitive way <strong>of</strong> classifying CBCs is to divide them<br />

into those in which round to oval (mononuclear) cells predominate and<br />

those in which segmented (polynuclear) cells predominate. However, this<br />

old method does allow the relevant differential diagnosis to be inferred<br />

from a cursory screening <strong>of</strong> a slide.<br />

Differential Diagnostic Notes<br />

Differential diagnosis when round to oval cells predominate<br />

➤ Reactive lymphocytoses and monocytoses, pp. 67 f., 89<br />

➤ Lymphatic system diseases (lymphomas, lymphocytic leukemia),<br />

p. 70<br />

➤ Acute leukemias, including episodes <strong>of</strong> chronic myeloid leukemia<br />

(CML), p. 96 ff.<br />

➤ Low counts <strong>of</strong> cells with segmented nuclei (agranulocytosis–<br />

aplastic anemia–myelodysplasia), pp. 86, 106, 148<br />

Differential diagnosis when segmented cells predominate<br />

➤ Reactive processes, p. 112 ff.<br />

➤ Chronic myeloid leukemia, p. 114 ff.<br />

➤ Osteomyelosclerosis, p. 122<br />

➤ Polycythemia vera, p. 162 ff.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Predominance <strong>of</strong> Mononuclear Round to Oval Cells<br />

Predominance <strong>of</strong> Mononuclear Round<br />

to Oval Cells (Table 5)<br />

63<br />

The cell counts (per microliter) show a wide range <strong>of</strong> values and depend<br />

on a variety <strong>of</strong> conditions even in normal blood. Any disease may therefore<br />

result in higher (leukocytosis) or lower cell counts (leukopenia). The<br />

assessment <strong>of</strong> cell counts requires the knowledge <strong>of</strong> normal values and<br />

their ranges (spreads) (Table 2, p. 12). The most important diagnostic indicator<br />

therefore is not the cell count but the cell type. In a first assessment,<br />

mononuclear (round to oval) and polynuclear (segmented) cells are<br />

compared. This is the first step in the differential diagnosis <strong>of</strong> the multifaceted<br />

spectrum <strong>of</strong> blood cells.<br />

Absolute or relative predominance <strong>of</strong> mononuclear cells points to a defined<br />

set <strong>of</strong> diagnostic probabilities. The differential diagnostic notes opposite<br />

can help with a first orientation.<br />

Consequently, the most important step is to distinguish between lymphatic<br />

cells and myeloid blasts. The nuclear morphology makes it possible<br />

to distinguish between the two cell types. In lymphatic cells the nucleus<br />

usually displays dense coarse chromatin with slate-like architecture and<br />

lighter zones between very dense ones. In contrast, the immature cells in<br />

myeloid leukemias contain nuclei with a more delicate reticular, sometimes<br />

sand-like chromatin structure with a finer, more irregular pattern.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


64<br />

Table 5 Diagnostic work-up for abnormalities in the white cell series with mononuclear<br />

cells predominating<br />

Clinical findings Hb MCH Leukocytes Segmented<br />

cells<br />

Fever, lymph nodes,<br />

possibly exanthema<br />

Fever, severe lymphoma,<br />

possibly<br />

spleen ↑<br />

Slowly progressing<br />

lymph node enlargement<br />

in several locations<br />

Night sweat, slowly<br />

progressing lymphoma<br />

(spleen),<br />

possibly fever<br />

Slowly progressing<br />

lymph node enlargement<br />

in one or a few<br />

locations<br />

Isolated severe<br />

splenomegaly<br />

Lymphocytes<br />

(%)<br />

n n ↓/n ↓ ↑<br />

Stimulated<br />

forms<br />

Other cells<br />

n n ↑ ↓ ↑ Large blastic<br />

stimulated cells<br />

(DD: lymphoblasts)<br />

↓/n ↓/n ↑ ↓ ↑↑ –<br />

↓ ↓ n/↑ ↓ ↑ Plasmacytoid<br />

cells, possibly<br />

rouleaux formation<br />

<strong>of</strong> the<br />

erythrocytes<br />

↓ n/↓ n/↑/↓ n/↓ Possibly ↑ Possibly cells<br />

with grooves<br />

↓ ↓ ↓/↑ ↓ Hairy cells<br />

Fever, angina n n ↓ ↓ ↑ Monocytes ↑<br />

Diffuse general<br />

symptoms<br />

Pale skin, fever<br />

(signs <strong>of</strong> hemorrhage)<br />

Fatigue, night<br />

sweat, possibly<br />

bone pain<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

↓ ↓ ↑ ↓ n Monocytes ↑<br />

↓ ↓ ↑/n/↓ ↓ ↓ Atypical round<br />

cells predominate<br />

↓ ↓ n/↓ n n Possibly rouleaux<br />

formations <strong>of</strong><br />

erythrocytes<br />

Diagnostic steps proceed from left to right. | The next step is usually unnecessary; optional<br />

step, may not progress the diagnosis; ⎯→ the next step is obligatory.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />


Thrombocytes<br />

Electrophoresis<br />

Tentative<br />

diagnosis<br />

n n/Y↑ Virus infection,<br />

e.g., rubeola<br />

n Y↑ Mononucleosis<br />

n/↓ n/Y↓ Leukemic<br />

non-Hodgkin<br />

lymphoma,<br />

esp. CLL<br />

n/↓ Possibly<br />

Y↑<br />

Immunocytoma<br />

(incl.<br />

Waldenströmdisease)<br />

n/↓ n Non-Hodgkin<br />

lymphoma,<br />

e.g. follicular<br />

lymphoma<br />

↓ n Hairy cell<br />

leukemia<br />

n n Agranulocytosis<br />

n Possibly<br />

a2↑<br />

Reactive<br />

monocytosis<br />

in chronic<br />

infection or<br />

tumor<br />

↓ n Acute<br />

leukemia<br />

n/↓ Sharp<br />

peaks ↑<br />

Predominance <strong>of</strong> Mononuclear Round to Oval Cells<br />

Multiple<br />

myeloma<br />

Evidence/advanced<br />

diagnostics<br />

Clinical developments,<br />

possibly serological<br />

tests<br />

Mononucleosis quick<br />

test, EBV serology<br />

(cytomegaly titer?)<br />

Marker analysis <strong>of</strong><br />

peripheral lymphocytes<br />

is sufficient in typical<br />

disease presentations;<br />

lymph node histology<br />

for treatment decisions,<br />

if necessary<br />

Immunoelectrophoresis;<br />

marker analysis for<br />

lymphocytes in blood<br />

and bone marrow;<br />

lymph node histology in<br />

case <strong>of</strong> doubt<br />

Lymph node histology<br />

Cell surface markers<br />

.<br />

Bone marrow<br />

Always<br />

involved in<br />

CLL, not<br />

always in<br />

other lymphomas<br />

Usually<br />

involved<br />

Ref. page<br />

p. 66<br />

p. 68<br />

p. 74<br />

p. 78<br />

.<br />

For the pur- p. 78<br />

pose <strong>of</strong> staging,<br />

possibly<br />

positive<br />

.<br />

Always p. 80<br />

involved, discrete<br />

in the<br />

beginning<br />

.<br />

⎯⎯⎯⎯⎯⎯⎯⎯→ Maturation<br />

arrest<br />

Determine disease<br />

origin<br />

Cytochemistry, marker<br />

analysis, possibly cytogenetics<br />

⎯⎯⎯⎯⎯⎯⎯⎯→<br />

Immunoelectrophoresis,<br />

skeletal X-ray<br />

⎯⎯⎯⎯⎯⎯⎯⎯→<br />

Marker for<br />

blasts<br />

Bone marrow<br />

contains<br />

plasma cells<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

p. 86<br />

p. 88<br />

p. 90ff<br />

p. 82<br />

65


66<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Reactive Lymphocytosis<br />

Lymphatic cells show wide variability and transform easily. This is usually<br />

seen as enlarged nuclei, a moderately loose, coarse chromatin structure,<br />

and a marked widening <strong>of</strong> the basophilic cytoplasmic layer.<br />

Clinical findings, which include acute fever symptoms, enlarged lymph<br />

nodes, and sometimes exanthema, help to identify a lymphatic reactive<br />

state. Unlike the case in acute leukemias, erythrocyte and thrombocyte<br />

counts are not significantly reduced. Although the granulocyte count is<br />

relatively reduced, its absolute value (per microliter) rarely falls below the<br />

lower limit <strong>of</strong> normal values.<br />

Morphologically normal lymphocytes predominate in the blood analyses in<br />

the following diseases:<br />

➤ Whooping cough (pertussis) with clear leukocytosis and total lymphocyte<br />

counts up to 20 000 and even 50 000/µl; occasionally, slightly<br />

plasmacytoid differentiation.<br />

➤ Infectious lymphocytosis, a pediatric infectious disease with fever <strong>of</strong><br />

short duration. Lymphocyte counts may increase to 50 000/µl.<br />

➤ Chickenpox, measles, and brucellosis, in which a less well-developed<br />

relative lymphocytosis is found, and the counts remain within the normal<br />

range.<br />

➤ Hyperthyroidism and Addison disease, which show relative lymphocytosis.<br />

➤ Constitutional relative lymphocytosis, which can reach up to 60% and<br />

occurs without apparent reason (mostly in asthenic teenagers).<br />

➤ Absolute granulocytopenias with relative lymphocytosis (p. 86).<br />

➤ Chronic lymphocytic leukemia (CLL), which is always accompanied by<br />

absolute and relative lymphocytosis, usually with high cell counts.<br />

Transformed, “stimulated” lymphocytes (“virocytes”) predominate in the<br />

CBC in the following diseases with reactive symptoms:<br />

➤ Lymphomatous toxoplasmosis does not usually involve significant<br />

leukocytosis. Slightly plasmacytoid cell forms are found.<br />

➤ In rubella infections the total leukocyte count is normal or low, and the<br />

lymphocytosis is only relatively developed. The cell morphology ranges<br />

from basophilic plasmacytoid cells to typical plasma cells.<br />

➤ In hepatitis the total leukocyte and lymphocyte counts are normal.<br />

However, the lymphocytes <strong>of</strong>ten clearly show plasmacytoid transformation.<br />

➤ The most extreme lymphocyte transformation is observed in mononucleosis<br />

(Epstein–Barr virus [EBV] or cytomegalovirus [CMV] infection)<br />

(p. 68).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

b<br />

d<br />

During lymphatic reactive states, variable cells with dense,<br />

round nuclei (e.g., virocytes) dominate the CBC<br />

Fig. 21 Lymphatic reactive states. a–e Wide variability <strong>of</strong> the lymphatic cells in a<br />

lymphotropic infection (in this case cytomegalovirus infection). Some <strong>of</strong> the cells<br />

may resemble myelocytes, but their chromatin is always denser than myelocyte<br />

chromatin.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

67<br />

c<br />

e


68<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Examples <strong>of</strong> Extreme Lymphocytic Stimulation:<br />

Infectious Mononucleosis<br />

Epstein–Barr virus infection should be considered when, after a prodromal<br />

fever <strong>of</strong> unknown origin, there are signs <strong>of</strong> enlarged lymph nodes and<br />

developing angina, and the blood analysis shows predominantly mononuclear<br />

cells and a slightly, or moderately, elevated leukocyte count. Varying<br />

proportions <strong>of</strong> the mononuclear cells (at least 20%) may be rather extensively<br />

transformed round cells (Pfeiffer cells, virocytes). Immunological<br />

markers are necessary to ascertain that these are stimulated lymphocytes<br />

(mostly T-lymphocytes) defending the B-lymphocyte stem population<br />

against the virus attack. The nuclei <strong>of</strong> these stimulated lymphocytes are<br />

two- to three-fold larger than those <strong>of</strong> normal lymphocytes and their<br />

chromatin has changed from a dense and coarse structure to a looser,<br />

more irregular organization. The cytoplasm is always relatively wide and<br />

more or less basophilic with vacuoles. Granules are absent. A small proportion<br />

<strong>of</strong> cells appear plasmacytoid. In the course <strong>of</strong> the disease, the<br />

degree <strong>of</strong> transformation and the proportions <strong>of</strong> the different cell morphologies<br />

change almost daily. A slight left shift and elevated monocyte<br />

count are <strong>of</strong>ten found in the granulocyte series.<br />

Acute leukemia is <strong>of</strong>ten considered in the differential diagnosis in addition<br />

to other viral conditions, because the transformed lymphocytes can<br />

resemble the blasts found in leukemia. Absence <strong>of</strong> a quantitative reduction<br />

<strong>of</strong> hematopoiesis in all the blood cell series, however, makes leukemia<br />

unlikely, as do the variety and speed <strong>of</strong> change in the cell morphology. Finally,<br />

serological tests (EBV antigen test, test for antibodies, and, if indicated,<br />

quick tests) can add clarification.<br />

Where serological tests are negative, the cause <strong>of</strong> the symptoms is usually<br />

cytomegalovirus rather than EBV.<br />

Characteristics <strong>of</strong> Infectious Mononucleosis<br />

Age <strong>of</strong> onset: School age<br />

Clinical findings: Enlarged lymph nodes (rapid onset), inflammations<br />

<strong>of</strong> throat and possibly spleen <br />

CBC: Leukocytes , stimulated lymph nodes, partially lymphoblasts<br />

(hematocrit and thrombocytes are normal)<br />

Further diagnostics: EBV serological test (IgM +), transaminases usually<br />

<br />

Differential diagnosis: Lymphomas (usually without fever), leukemia<br />

(usually Hb , thrombocytes ) Persistent disease (more than 3<br />

weeks): possibly test for blood cell markers, lymph node cytology<br />

( histology)<br />

Course, treatment: Spontaneous recovery within 2–4 weeks; general<br />

palliation<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

Extreme transformation <strong>of</strong> lymphocytes leads to blast-like cells:<br />

mononucleosis<br />

c<br />

d<br />

Fig. 22 Lymphocytes during viral infection. a “Blastic,” lymphatic reactive form<br />

(Pfeiffer cell), in addition to less reactive virocytes in Epstein–Barr virus (EBV) infection.<br />

This phase with blastic cells lasts only a few days. b Virocyte (1) with homogeneous<br />

deep blue stained cytoplasm in EBV infection, in addition to normal<br />

lymphocyte (2) and monocyte (3). c Virus infection can also lead to elevated<br />

counts <strong>of</strong> large granulated lymphocytes (LGL) (1). Monocyte (2). d Severe<br />

lymphatic stress reaction with granulated lymphocytes. A lymphoma must be<br />

considered if this finding persists.<br />

69<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

b


70<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Diseases <strong>of</strong> the Lymphatic System<br />

(Non-Hodgkin Lymphomas)<br />

Malignant diseases <strong>of</strong> the lymphatic system are further classified as Hodgkin<br />

and non-Hodgkin lymphomas (NHL). NHL will be discussed here because<br />

most NHLs can be diagnosed on the blood analysis.<br />

➤ Non-Hodgkin lymphomas arise mostly from small or blastic B-cells.<br />

➤ Small-cell NHL cells are usually leukemic and relatively indolent, <strong>of</strong> the<br />

type <strong>of</strong> chronic lymphocytic leukemia and its variants.<br />

➤ Blastic NHL (precursor lymphoma) is not usually leukemic. An exception<br />

is the lymphoblastic lymphoma, which takes its course as an acute<br />

lymphocytic leukemia.<br />

➤ Plasmacytoma is an osteotropic B-cell lymphoma that releases not its<br />

cells but their products (immunoglobulins) into the bloodstream.<br />

➤ Malignant lymphogranulomatosis (Hodgkin disease) cannot be diagnosed<br />

on the basis <strong>of</strong> blood analysis. It is therefore discussed under<br />

lymph node cytology (p. 176).<br />

The modern pathological classification <strong>of</strong> lymphomas is based on morphology<br />

and cell immunology. The updated classification system according<br />

to Lennert (Kiel) has been adopted in Germany and was recently modified<br />

to reflect the WHO classification. The definitions <strong>of</strong> stages I–IV in NHL<br />

agree with the Ann Arbor classification for Hodgkin’s disease.<br />

Table 6a Classification <strong>of</strong> non-Hodgkin: comparison <strong>of</strong> the relevant classes in the Kiel and WHO classifications<br />

WHO Kiel Clinical<br />

Characteristics<br />

Precursor lymphomas/leukemias<br />

➤ Precursor-B-lymphoblastic<br />

lymphoma<br />

➤ Precursor B-cell active lymphoblastic<br />

leukemia<br />

Mature B-cell lymphoma<br />

➤ Chronic lymphocytic leukemia<br />

– contains the lymphoplasmacytoid<br />

immunocytoma<br />

B-lymphoblastic<br />

lymphoma<br />

B-ALL<br />

B-CLL<br />

Lymphoplasmacytoid<br />

immunocytoma<br />

Marker*<br />

Aggressive Tdt,<br />

CD19, 22,<br />

79 a<br />

Usually indolent<br />

Usually indolent<br />

➤ B-cell prolymphocytic leukemia Aggressive<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

CD5, 19,<br />

20, 23<br />

tris 12 or<br />

13 q-,<br />

11 q-, 6 q-,<br />

p 53


Table 6a Continued<br />

WHO Kiel Clinical<br />

Characteristics<br />

➤ Lymphoplasmacytic leukemia Lymphoplasmacytic<br />

immunocytoma<br />

Sometimes IgM<br />

paraprotein<br />

(Waldenström<br />

disease)<br />

➤ Mantle cell lymphoma Centrocytic lymphoma Usually aggressive<br />

➤ Marginal zone lymphoma<br />

– Nodal<br />

– Extranodal in mucosa (MALT)<br />

– Lienal (splenic)<br />

➤ Follicular lymphoma<br />

– Grade 1, 2<br />

– Grade 3 (a, b)<br />

Monocytoid lymphoma<br />

MALT (mucosa-assoc.<br />

lymphoid tissue) lymphoma<br />

Lymphoma with<br />

splenomegaly<br />

Centroblastic/centrocytic<br />

lymphoma;<br />

centroblastic lymphoma<br />

(a), secondary (b), follicular<br />

Usually aggressive<br />

Often indolent<br />

Usually indolent<br />

Highly malignant<br />

Marker*<br />

–/+ CD5<br />

– CD23,<br />

CD5<br />

t(11;14)<br />

bcl-1<br />

rearr.<br />

–CD5,<br />

CD23<br />

–CD5,<br />

CD10<br />

t(14;18)<br />

bcl2<br />

➤ Hairy cell leukemia Hairy cell leukemia Usually indolent –CD103,<br />

CD11 c,<br />

CD25<br />

t(2;8)<br />

t(8;14)<br />

➤ Plasma cell myeloma (plasmacytoma)<br />

– Monoclonal hypergammaglobulinemia<br />

(GUS)<br />

– Solitary bone plasmacytoma<br />

– Extraosseous bone plasmacytoma<br />

– Primary amyloidosis<br />

– Heavy-chain disease<br />

Primary large-cell lymphoma<br />

➤ Diffuse large-cell B-cell<br />

lymphoma<br />

– Centroblastic<br />

– Immunoblastic<br />

– Large-cell anaplastic<br />

Predominance <strong>of</strong> Mononuclear Round to Oval Cells<br />

[Plasmacytoma is not<br />

included in the Kiel<br />

classification]<br />

Centroblastic<br />

Immunoblastic<br />

Large-cell anaplastic<br />

Extremely malignant<br />

Extremely malignant<br />

Extremely malignant<br />

➤ Burkitt lymphoma Burkitt lymphoma Extremely malignant<br />

* Cited markers are positive, absent markers are indicated with a minus sign.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

CD 138<br />

CD20,<br />

79 a, 19,<br />

22<br />

t(2;8)<br />

t(8;14)<br />

myc<br />

71


72<br />

Table 6b T-cell lymphomas (since T-cell lymphomas make up only 10% <strong>of</strong> all NHLs, this table gives just a brief<br />

characterization; for markers see Table 7)<br />

WHO<br />

( Kiel)<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Classification Manifestation<br />

➤ T-prolymphocytic leukemia<br />

➤ Large granular lymphocyte leukemia (LGL)<br />

➤ T-cell lymphoblastic leukemia<br />

Leukemic<br />

Leukemic<br />

Leukemic<br />

Clinical characteristics<br />

Aggressive<br />

Sometimes indolent<br />

Aggressive<br />

➤ Sézary syndrome, mycosis fungoides Cutaneous Chronic, progressive<br />

➤ Angioimmunoblastic T-cell lymphoma (AILD) Nodal and ENT Usually aggressive<br />

➤ Lymphoblastic T-cell lymphoma<br />

➤ T-cell zone lymphoma (nonspecific peripheral<br />

lymphoma)<br />

➤ Lennert lymphoma with multifocal epithelioid<br />

cells<br />

➤ Large-cell anaplastic lymphoma (ki1)<br />

Nodal<br />

Nodal<br />

Nodal<br />

Nodal<br />

Differentiation <strong>of</strong> the Lymphatic Cells and Cell Surface<br />

Marker Expression in Non-Hodgkin Lymphoma Cells<br />

Aggressive<br />

Sometimes slowly<br />

progressive<br />

Sometimes slowly<br />

progressive<br />

Aggressive<br />

Non-Hodgkin lymphoma cells derive monoclonally from specific stages in<br />

the B- or T-cell differentiation, and their surface markers reflect this. The<br />

surface markers are identified in immunocytological tests (Table 7) carried<br />

out on heparinized blood or bone marrow spicules.<br />

The blastic lymphomas will not be discussed further in the context <strong>of</strong><br />

diagnostics based on blood cell morphology. The findings in the primarily<br />

leukemic forms <strong>of</strong> the disease, such as lymphoblastic lymphoma, resemble<br />

those for ALL (p. 104). Other blastic lymphomas can usually only<br />

be diagnosed on the basis <strong>of</strong> lymph node tissue (Fig. 65). Of course, despite<br />

all the progress in the analysis <strong>of</strong> blood cell differentiation, <strong>of</strong>ten analysis<br />

<strong>of</strong> histological slides in conjunction with the blood analysis is required for<br />

a confident diagnosis.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Table 7 Cell surface markers <strong>of</strong> lymphatic cells in leukemic, low-grade malignant non-Hodgkin lymphoma<br />

Marker B-CLL B-PLL HCL FL MCL SLVL T-CLL GL SS T-PLL ATLL<br />

Predominance <strong>of</strong> Mononuclear Round to Oval Cells<br />

S Ig (+) ++ ++ ++ ++ ++ – – – – –<br />

CD 2 – – – – – – + + + + +<br />

CD 3 – – – – – – + – + + +<br />

CD 4 – – – – – – – – + +/– +<br />

CD 5 ++ – – – + – – – + + +<br />

CD 7 – – – – – – – – +/– + –/+<br />

CD 8 – – – – – – + +/– +/– +/– +/–<br />

CD 19/20/24 ++ ++ ++ + + ++ – – – – –<br />

CD 22 +/– ++ ++ + + ++ – – – – –<br />

CD 10 – – – +/– – – – – – – –<br />

CD 25 – – ++ – – +/– – – – – +<br />

CD 56 – – – – – – – + – – –<br />

CD 103 – – ++ – – – – – – – –<br />

CLL chronic lymphocytic leukemia; PLL prolymphocytic leukemia; HCL hairy cell leukemia; FL follicular lymphoma; MCL mantle cell lymphoma; SLVL splenic lymphoma with villous lymphocytes;<br />

LGL large granular lymphocyte leukemia; SS Sézary syndrome; ATLL adult T-cell lymphoma.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

73


74<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Chronic Lymphocytic Leukemia (CLL) and Related Diseases<br />

A chronic lymphadenoma, or chronic lymphocytic leukemia, can sometimes<br />

be clinically diagnosed with some certainty. An example is the case<br />

<strong>of</strong> a patient (usually older) with clearly enlarged lymph nodes and significant<br />

lymphocytosis (in 60% <strong>of</strong> the cases this is greater than 20 000/µl and<br />

in 20% <strong>of</strong> the cases it is greater than 100 000/µl) in the absence <strong>of</strong> symptoms<br />

that point to a reactive disorder. The lymphoma cells are relatively<br />

small, and the nuclear chromatin is coarse and dense. The narrow layer <strong>of</strong><br />

slightly basophilic cytoplasm does not contain granules. Shadows around<br />

the nucleus are an artifact produced by chromatin fragmentation during<br />

preparation (Gumprecht’s nuclear shadow). In order to confirm the diagnosis,<br />

the B-cell markers on circulating lymphocytes should be characterized<br />

to show that the cells are indeed monoclonal. The transformed<br />

lymphocytes are dispersed at varying cell densities throughout the bone<br />

marrow and the lymph nodes. A slowly progressing hypogammaglobulinemia<br />

is another important indicator <strong>of</strong> a B-cell maturation disorder.<br />

Transition to a diffuse large-cell B-lymphoma (Richter syndrome) is<br />

rare: B-prolymphocytic leukemia (B-PLL) displays unique symptoms. At<br />

least 55% <strong>of</strong> the lymphocytes in circulating blood have large central<br />

vacuoles. When 15–55% <strong>of</strong> the cells are prolymphocytes, the diagnosis <strong>of</strong><br />

atypical CLL, or transitional CLL/PLL is confirmed. In some CLL-like diseases,<br />

the layer <strong>of</strong> cytoplasm is slightly wider. B-CLL was defined as lymphoplasmacytoid<br />

immunocytoma in the Kiel classification. According to<br />

the WHO classification, it is a B-CLL variation (compare this with lymphoplasmacytic<br />

leukemia, p. 78). CLL <strong>of</strong> the T-lymphocytes is rare. The cells<br />

show nuclei with either invaginations or well-defined nucleoli (T-prolymphocytic<br />

leukemia). The leukemic phase <strong>of</strong> cutaneous T-cell lymphoma<br />

(CTCL) is known as Sézary syndrome. The cell elements in this syndrome<br />

and T-PLL are similar.<br />

Fig. 23 CLL. a Extensive proliferation <strong>of</strong> lymphocytes with densely structured<br />

nuclei and little variation in CLL. Nuclear shadows are frequently seen, a sign <strong>of</strong><br />

the fragility <strong>of</strong> the cells (magnification 400). b Lymphocytes in CLL with typical<br />

coarse chromatin structure and small cytoplasmic layer (enlargement <strong>of</strong> a section<br />

from 23 a, magnification 1000); only discreet nucleoli may occur. c Slightly<br />

eccentric enlargement <strong>of</strong> the cytoplasm in the lymphoplasmacytoid variant <strong>of</strong><br />

CLL.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

d<br />

Monotonous proliferation <strong>of</strong> small lymphocytes suggests chronic<br />

lymphocytic leukemia (CLL)<br />

Fig. 23 d Proliferation <strong>of</strong> atypical large lymphocytes (1) with irregularly structured<br />

nucleus, well-defined nucleolus, and wide cytoplasm (atypical CLL or transitional<br />

form CLL/PLL). e Bone marrow cytology in CLL: There is always strong<br />

proliferation <strong>of</strong> the typical small lymphocytes, which are usually spread out diffusely.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

75<br />

b<br />

e


76<br />

Table 8 Staging <strong>of</strong> CLL according to Rai (1975)<br />

Stage Identifying criteria/definition<br />

(Low risk) 0 Lymphocytosis 15 000/µl<br />

Bone marrow infiltration 40%<br />

(Intermediate<br />

risk)<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

I<br />

II<br />

Lymphocytosis and lymphadenopathy<br />

Lymphocytosis and hepatomegaly and/or splenomegaly<br />

(with or without lymphadenopathy)<br />

(High risk) III Lymphocytosis and anemia (Hb 11.0 g/dl) (with or<br />

without lymphadenopathy and/or organomegaly)<br />

IV Lymphocytosis and thrombopenia ( 100 000/µl)<br />

(with or without anemia, lymphadenopathy, or<br />

organomegaly)<br />

Table 9 Staging <strong>of</strong> CLL according to Binet (1981)<br />

Stage Identifying criteria/definition<br />

A Hb 10.0 g/dl, normal thrombocyte count<br />

3 regions with enlarged lymph nodes<br />

B Hb 10.0 g/dl, normal thrombocyte count<br />

3 regions with enlarged lymph nodes<br />

C Hb 10.0 g/dl and/or thrombocyte count 100 000/µl<br />

independent <strong>of</strong> the number <strong>of</strong> affected locations<br />

Characteristics <strong>of</strong> CLL<br />

Age <strong>of</strong> onset: Mature adulthood<br />

Clinical presentation: Gradual enlargement <strong>of</strong> all lymph nodes, usually<br />

moderately enlarged spleen, slow onset <strong>of</strong> anemia and increasing<br />

susceptibility to infections, later thrombocytopenia<br />

CBC: In all cases absolute lymphocytosis; in the course <strong>of</strong> the disease<br />

Hb , thrombocytes , immunoglobulin <br />

Further diagnostics: Lymphocyte surface markers (see pp. 68 ff.); bone<br />

marrow (always infiltrated); lymph node histology further clarifies<br />

the diagnosis<br />

Differential diagnosis: (a) Related lymphomas: marker analysis,<br />

lymph node histology; (b) acute leukemia: cell surface marker analysis,<br />

cytochemistry, cytogenetics (pp. 88 ff.)<br />

Course, therapy: Individually varying, usually fairly indolent course;<br />

in advanced stages or fast progressing disease: moderate chemotherapy<br />

(cell surface marker, see Table 7)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Atypical lymphocytes are not part <strong>of</strong> B-CLL<br />

Fig. 24 Lymphoma <strong>of</strong> the B-cell and T-cell lineages. a Prevalence <strong>of</strong> large lymphocyteswithclearlydefinednucleoliandwidecytoplasm:prolymphocyticleukemia<strong>of</strong><br />

the B-cell series (B-PLL). b The presence <strong>of</strong> large blastic cells (arrow) in CLL suggest a<br />

rare transformation (Richter syndrome). c The rare Sézary syndrome (T-cell lymphoma<br />

<strong>of</strong> the skin) is characterized by irregular, indented lymphocytes. d Prolymphocytic<br />

leukemia <strong>of</strong> the T-cell series (T-PLL) with indented nuclei and nucleoli<br />

(rare). e Bone marrow in lymphoplasmacytic immunocytoma: focal or diffuse lymphocyte<br />

infiltration (e.g., 1), plasmacytoid lymphocytes (e.g., 2) and plasma cells<br />

(e.g., 3). Red cell precursors predominate (e.g., basophilic erythroblasts, arrow).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

77<br />

b<br />

d<br />

e


78<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

The pathological staging for CLL is always Ann Arbor stage IV because the<br />

bone marrow is affected. In the classifications <strong>of</strong> disease activity by Rai<br />

and Binet (analogous to that for leukemic immunocytoma), the transition<br />

between stages is smooth (Tables 8 and 9).<br />

Lymphoplasmacytic Lymphoma<br />

The CBC shows lymphocytes with relatively wide layers <strong>of</strong> cytoplasm. The<br />

bone marrow contains a mixture <strong>of</strong> lymphocytes, plasmacytic lymphocytes,<br />

and plasma cells. In up to 30% <strong>of</strong> cases paraprotein is secreted, predominantly<br />

monoclonal IgM. This constitutes the classic Waldenström<br />

syndrome (Waldenström macroglobulinemia). The differential diagnosis<br />

may call for exclusion <strong>of</strong> the rare plasma cell leukemia (see p. 82) and <strong>of</strong><br />

lymphoplasmacytoid immunocytoma, which is closely related to CLL (see<br />

p. 74).<br />

Characteristics<br />

➤ Lymphoplasmacytoid immunocytoma: This is a special form <strong>of</strong> B-<br />

CLL in which usually only a few precursors migrate into the bloodstream<br />

(a lesser degree <strong>of</strong> malignancy). A diagnosis may only be<br />

possible on the basis <strong>of</strong> bone marrow or lymph node analysis.<br />

➤ Lymphoplasmacytic lymphoma: Few precursors migrate into the<br />

bloodstream (i.e., bone marrow or lymph node analysis is sometimes<br />

necessary). There is <strong>of</strong>ten secretion <strong>of</strong> IgM paraprotein,<br />

which can lead to hyperviscosity.<br />

Further diagnostics: Marker analyses in circulating cells, lymph node cytology,<br />

bone marrow cytology and histology, and immunoelectrophoresis.<br />

Plasmacytoma cells migrate into the circulating blood in appreciable<br />

numbers in only 1–2% <strong>of</strong> all cases <strong>of</strong> plasma cell leukemia. Therefore, paraproteins<br />

must be analyzed in bone marrow aspirates (p. 82).<br />

Facultative Leukemic Lymphomas<br />

(e.g., Mantle Cell Lymphoma and Follicular Lymphoma)<br />

In all cases <strong>of</strong> non-Hodgkin lymphoma, the transformed cells may migrate<br />

into the blood stream. This is usually observed in mantle cell lymphoma:<br />

The cells are typically <strong>of</strong> medium size. On close examination, their nuclei<br />

show loosely structured chromatin and they are lobed with small indentations<br />

(cleaved cells). Either initially, or, more commonly, during the course<br />

<strong>of</strong> the disease, a portion <strong>of</strong> cells becomes larger with relatively enlarged<br />

nuclei (diameter 8–12 µm). These larger cells are variably “blastoid.” Lymphoid<br />

cells also migrate into the blood in stage IV follicular lymphoma.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Deep nuclear indentation suggests follicular lymphoma or<br />

mantle cell lymphoma<br />

a b<br />

Fig. 25 Mantle cell lymphoma. a Fine, dense chromatin and small indentations<br />

<strong>of</strong> the nuclei suggest migration <strong>of</strong> leukemic mantle cell lymphoma cells into the<br />

blood stream. b Denser chromatin and sharp indentations suggest migration <strong>of</strong><br />

follicular lymphoma cells into the blood stream. c Diffuse infiltration <strong>of</strong> the bone<br />

marrow with polygonal, in some cases indented lymphatic cells in mantle cell<br />

lymphoma. Bone marrow involvement in follicular lymphoma can <strong>of</strong>ten only be<br />

demonstrated by histological and cytogenetic studies.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

79<br />

c


80<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

“Monocytoid” cells with a wide layer <strong>of</strong> only faintly staining cytoplasm<br />

occur in blood in marginal zone lymphadenoma (differential diagnosis:<br />

lymphoplasmacytic immunocytoma).<br />

Lymphoma, Usually with Splenomegaly<br />

(e.g., Hairy Cell Leukemia and Splenic Lymphoma<br />

with Villous Lymphocytes)<br />

Hairy cell leukemia (HCL). In cases <strong>of</strong> slowly progressive general malaise<br />

with isolated splenomegaly and pancytopenia revealed by CBC (leukocytopenia,<br />

anemia, and thrombocytopenia), the predominating mononuclear<br />

cells deserve particular attention. The nucleus is oval, <strong>of</strong>ten kidney beanshaped,<br />

and shows a delicate, elaborate chromatin structure. The cytoplasm<br />

is basophilic and stains slightly gray. Long, very thin cytoplasmic<br />

processes give the cells the hairy appearance that gave rise to the term<br />

“hairy cell leukemia” used in the international literature. The disease affects<br />

the spleen, liver, and bone marrow. Severe lymphoma is usually absent.<br />

Aside from the typical hairy cells with their long, thin processes,<br />

there are also cells with a smooth plasma membrane, similar to cells in immunocytoma.<br />

A variant shows well-defined nucleoli (HCL-V, hairy cell<br />

leukemia variant). A bone marrow aspirate <strong>of</strong>ten does not yield material<br />

for an analysis (“punctio sicca” or “empty tap”) because the marrow is very<br />

fibrous. Apart from the bone marrow histology, advanced cell diagnostics<br />

are therefore very important, in particular in the determination <strong>of</strong> blood<br />

cell surface markers (immunophenotyping). This analysis reveals CD 103<br />

and 11 c as specific markers and has largely replaced the test for tartrateresistant<br />

acid phosphatase.<br />

Splenic lymphoma with villous lymphocytes (SLVL). This lymphatic system<br />

disease mostly affects the spleen. There is little involvement <strong>of</strong> the bone<br />

marrow and no involvement <strong>of</strong> the lymph nodes. The blood contains lymphatic<br />

cells, which resemble hairy cells. However, the “hairs,” i.e., cytoplasmic<br />

processes, are thicker and mostly restricted to one area at the cell<br />

pole, and the CD 103 marker is absent.<br />

Splenomegaly may develop in all non-Hodgkin lymphomas. In hairy cell<br />

leukemia, the rare splenic lymphadenoma with villous lymphocytes<br />

(SLVL) and marginal zone lymphadenoma may be seen. These mostly affect<br />

the spleen.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Cytoplasmic processes the main feature <strong>of</strong> hairy cell leukemia<br />

Fig. 26 Hairy cell leukemia and splenic lymphoma. a and b Ovaloid nuclei and<br />

finely “fraying” cytoplasm are characteristics <strong>of</strong> cells in hairy cell leukemia (HCL).<br />

c Occasionally, the hairy cell processes appear merely fuzzy. d and e When the cytoplasmic<br />

processes look thicker and much less like hair, diagnosis <strong>of</strong> the rare splenic<br />

lymphoma with villous lymphocytes (SLVL) must be considered. Here, too, the<br />

next diagnostic step is analysis <strong>of</strong> cell surface markers.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

81<br />

b<br />

d<br />

e


82<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Monoclonal Gammopathy (Hypergammaglobulinemia),<br />

Multiple Myeloma*, Plasma Cell Myeloma, Plasmacytoma<br />

Plasmacytoma is the result <strong>of</strong> malignant transformation <strong>of</strong> the most<br />

mature B-lymphocytes (Fig. 1, p. 2). For this reason the diagnostics <strong>of</strong> this<br />

disease will be discussed here, even though migration <strong>of</strong> its specific cells<br />

into the blood stream (plasma cell leukemia) is extremely rare (1–2%).<br />

Immunoelectrophoresis <strong>of</strong> serum and urine is performed when electrophoresis<br />

shows very discrete gammaglobulin, or globulin, fractions, or<br />

when hypogammaglobulinemia is found (in light-chain plasmacytoma). A<br />

wide range <strong>of</strong> possibilities arises for the differential diagnosis <strong>of</strong> monoclonal<br />

transformed cells (Table 10).<br />

The presence <strong>of</strong> more than 10% <strong>of</strong> plasma cells, or atypical plasma cells<br />

in the bone marrow, is an important diagnostic factor in the diagnosis <strong>of</strong><br />

plasmacytoma. For more criteria, see p. 84.<br />

Table 10 Differential diagnosis <strong>of</strong> monoclonal hypergammaglobulinemia<br />

Type Characteristics<br />

Benign disorders<br />

➤ Essential hypergammaglobulinemia<br />

= MGUS (monoclonal gammopathy<br />

<strong>of</strong> unknown significance)<br />

➤ Symptomatic hypergammaglobulinemia,<br />

secondary to<br />

– Infections<br />

– Tumors<br />

– Autoimmune disease<br />

Malignant diseases<br />

➤ Plasmocytoma<br />

(usually IgG, A or light-chain<br />

[Bence Jones protein], rarely IgM,<br />

D, E) 90% disseminated (multiple<br />

myeloma), 5% solitary, 5%<br />

extramedullary (like a lymphoma<br />

or ENT tumor)<br />

➤ Lymphoma<br />

e. g., immunocytoma, CLL<br />

(potentially all lymphomas <strong>of</strong> the<br />

B-cell series)<br />

Usually in advanced age<br />

10%, plasma cells found in the<br />

bone marrow, no progression,<br />

normal polyclonal Ig<br />

All ages (otherwise as above)<br />

– Osteolysis or X-ray with severe<br />

osteoporosis<br />

– Plasmocytosis <strong>of</strong> the bone marrow<br />

10%<br />

– Monoclonal gammaglobulin in<br />

serum/urine with progression<br />

– Enlarged lymph nodes<br />

– Usually blood lymphocytosis<br />

– Monoclonal immunoglobulin,<br />

usually IgM<br />

* The current WHO classification suggests “multiple myeloma” (MM) for generalized<br />

plasmacytoma and “plasmacytoma” only for the rare solitary or nonosseous form <strong>of</strong><br />

plasmacytoma.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Plasmacytoma cannot be diagnosed without bone marrow analysis<br />

Fig. 27 Reactive plasmacytosis and plasmacytoma. a Bone marrow cytology<br />

with clear reactive features in the granulocyte series: strong granulation <strong>of</strong> promyelocytes<br />

(1) and myelocytes (2), eosinophilia (3), and plasma cell proliferation<br />

(4): reactive plasmacytosis (magnification 630). b Extensive (about 50%) infiltration<br />

<strong>of</strong> the bone marrow <strong>of</strong> mostly well-differentiated plasma cells: multiple<br />

myeloma (magnification 400).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

83<br />

a<br />

b


84<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Variability <strong>of</strong> Plasmacytoma Morphology<br />

It is not easy to visually distinguish malignant cells from normal plasma<br />

cells. Like lymphocytes, normal plasma cells have a densely structured nucleus.<br />

Plasma cell nuclei with radial chromatin organization, known as<br />

“wheel-spoke nuclei,” are mostly seen during histological analysis.<br />

The following attributes suggest a malignant character <strong>of</strong> plasma cells:<br />

the cells are unusually large (Fig. 28 c), they contain crystalline inclusions<br />

or protein inclusions (“Russell bodies”) (Fig. 28 b), or they have more than<br />

one nucleus (Fig. 28 c).<br />

In the differential diagnosis, they must be distinguished from hematopoietic<br />

precursor cells (Fig. 28 d), osteoblasts (Fig. 20 c), and blasts in acute<br />

leukemias (see Fig. 31, p. 97).<br />

Bone marrow involvement may be focal or in rare cases even solitary.<br />

Aside from cytological tests, bone marrow histology assays are therefore<br />

indicated. Sometimes, the biopsy must be obtained from a clearly identified<br />

osteolytic region.<br />

Although plasmacytomas progress slowly, staging criteria are available<br />

(staging according to Salmon and Durie) (Table 11).<br />

Therapy may be put on hold in stage I. Smoldering indolent myeloma<br />

can be left for a considerable time without the introduction <strong>of</strong> therapy<br />

stress. However, chemotherapy is indicated once the myeloma has<br />

exceeded the stage 1 criteria.<br />

Table 11 Staging <strong>of</strong> plasmacytomas according to Salmon and Durie<br />

Stage I<br />

All the following are present:<br />

– Hb 10 g/dl<br />

– Serum calcium is normal<br />

– X-ray shows normal bone structure<br />

or solitary skeletal plasmacytoma<br />

– IgG 5 g/dl*<br />

IgA 3 g/dl*<br />

Light chains in the urine*<br />

4 g/24 h<br />

* Monoclonal in each case.<br />

Stage II<br />

Findings fulfill neither stage I nor<br />

stage III criteria<br />

Stage III<br />

One or more <strong>of</strong> the following are<br />

present:<br />

– Hb 8.5 g/dl<br />

– Serum calcium is elevated<br />

– X-ray shows advanced bone lesions<br />

– IgG 7 g/dl*<br />

IgA 5 g/dl*<br />

Light chains in the urine:<br />

12 g/24 h<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Atypias and differential diagnoses <strong>of</strong> multiple myeloma<br />

Fig. 28 Atypical cells in multiple myeloma. a Extensive infiltration <strong>of</strong> the bone<br />

marrow by loosely structured, slightly dedifferentiated plasma cells with wide cytoplasm<br />

in multiple myeloma. b In multiple myeloma, vacuolated cytoplasmic<br />

protein precipitates (Russell bodies) may be seen in plasma cells but are without<br />

diagnostic significance. c Binuclear plasma cells are frequently observed in multiple<br />

myeloma (1). Mitotic red cell precursor (2). d Differential diagnosis: red cell<br />

precursor cells can sometimes look like plasma cells. Proerythroblast (1) and basophilic<br />

erythroblast (2).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

85<br />

b<br />

d


86 Abnormalities <strong>of</strong> the White Cell Series<br />

Relative Lymphocytosis Associated with<br />

Granulocytopenia (Neutropenia) and Agranulocytosis<br />

Neutropenia is defined by a decrease in the number <strong>of</strong> neutrophilic<br />

granulocytes with segmented nuclei to less than 1500/µl (1.5 10 9 /l). A<br />

neutrophil count <strong>of</strong> less than 500/µl (0.5 10 9 /l) constitutes agranulocytosis.<br />

Absolute granulocytopenias with benign cause develop into relative<br />

lymphocytoses.<br />

In the most common clinical picture, drug-induced acute agranulocytosis,<br />

the bone marrow is either poor in cells or lacks granulopoietic precursor<br />

cells (aplastic state), or shows a “maturation block” at the myeloblast–promyelocyte<br />

stage. The differential diagnosis <strong>of</strong> pure agranulocytosis<br />

versus aplasias <strong>of</strong> several cell lines is outlined on page 146.<br />

Classification <strong>of</strong> Neutropenias and Agranulocytoses<br />

1. Drug-induced:<br />

a) Dose-independent—acute agranulocytosis. Caused by hypersensitivity<br />

reactions: for example to pyrazolone, antirheumatic drugs<br />

(anti-inflammatory agents), antibiotics, or thyrostatic drugs.<br />

b) Relatively dose-dependent—subacute agranulocytosis (observed for<br />

carbamazepine [Tegretol], e.g. antidepressants, and cytostatic<br />

drugs).<br />

c) Dose-dependent—cytostatic drugs, immunosuppressants.<br />

2. Infection-induced:<br />

a) E.g., EBV, hepatitis, typhus, brucellosis.<br />

3. Autoimmune neutropenia:<br />

a) With antibody determination <strong>of</strong> T-cell or NK-cell autoimmune response<br />

b) In cases <strong>of</strong> systemic lupus erythematosus (SLE), Pneumocystis carinii<br />

pneumonia (PCP), Felty syndrome<br />

c) In cases <strong>of</strong> selective hypoplasia <strong>of</strong> the granulocytopoiesis (“pure<br />

white cell aplasia”)<br />

4. Congenital and familial neutropenias:<br />

Various pediatric forms; sometimes not expressed until adulthood,<br />

e.g. cyclical neutropenia<br />

5. Secondary neutropenia in bone marrow disease:<br />

Myelodysplastic syndromes (MDS), e.g., acute leukemia, plasmacytoma,<br />

pernicious anemia<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Bone marrow diagnosis is indicated in cases <strong>of</strong> unexplained agranulocytosis<br />

Fig. 29 The bone marrow in agranulocytosis. a In the early phase <strong>of</strong> agranulocytosis<br />

the bone marrow shows only red cell precursor cells (e.g., 1), plasma cells<br />

(2), and lymphocytes (3); in this sample a myeloblast—a sign <strong>of</strong> regeneration—is<br />

already present (4). b Bone marrow in agranulocytosis during the promyelocytic<br />

phase, showing almost exclusively promyelocytes (e.g., 1); increased eosinophilic<br />

granulocytes (2) are also present.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

87<br />

a<br />

b


88<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Monocytosis<br />

If mononuclear cells stand out in showing an unusually elaborate nuclear<br />

structure with ridges and lobes and a wider cytoplasmic layer with very<br />

delicate granules (for characteristics see p. 46, for cell function, see p. 6),<br />

and this is in the context <strong>of</strong> relative ( 10%) or absolute monocytosis (cell<br />

count 900/µl), a series <strong>of</strong> possible triggers must be considered (Table<br />

12). If the morphology does not clearly identify the cells as monocytes,<br />

then esterase assays should be done in a hematological laboratory using<br />

unstained smears.<br />

Table 12 Possible causes <strong>of</strong> monocytosis<br />

Infection<br />

Nonspecific monocytosis occurs in<br />

many bacterial infections during<br />

recuperation from or in the chronic<br />

phase <strong>of</strong>:<br />

– Mononucleosis (aside from stimulated<br />

lymphocytes there are also<br />

monocytes)<br />

– Listeriosis<br />

– Acute viral hepatitis<br />

– Parotitis epidemica (mumps)<br />

– Chickenpox<br />

– Recurrent fever<br />

– Syphilis<br />

– Tuberculosis<br />

– Endocarditis lenta<br />

– Brucellosis (Bang disease)<br />

– Variola vera (smallpox)<br />

– Rocky mountain spotted fever<br />

– Malaria<br />

– Paratyphoid fever<br />

– Kala-azar<br />

– Thypus fever<br />

– Trypanosomiasis (African sleeping<br />

sickness)<br />

Chronic reactive condition <strong>of</strong> the<br />

immune system, e. g., in:<br />

– Autoimmune diseases<br />

– Chronic dermatoses<br />

– Regional ileitis<br />

– Sarcoidosis<br />

Paraneoplasm<br />

(as attempted immune defense),<br />

e. g., in:<br />

– Large solid tumors<br />

– Lymphogranulomatosis<br />

Neoplasm<br />

– Chronic myelomonocytic<br />

leukemia (CMML, p. 107)<br />

– Acute monocytic leukemia<br />

(p. 100)<br />

– Acute myelomonocytic leukemia<br />

(p. 98)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Conspicuously large numbers <strong>of</strong> monocytes usually indicate a<br />

reactive disorder. Only a monotonous predominance <strong>of</strong> monocytes<br />

suggests leukemia<br />

a b<br />

Fig. 30 Reactive monocytosis and monocytic leukemia. a Reactive and neoplastic<br />

monocytes are morphologically indistinguishable; here two relatively condensed<br />

monocytes in reactive monocytosis are shown. b Whenever monocytes are<br />

found exclusively, a malignant etiology is likely: in this case AML M5 b according to<br />

the FAB classification (see p. 100). Auer bodies (arrow). c Monocytes <strong>of</strong> different<br />

degrees <strong>of</strong> maturity, segmented neutrophilic granulocytes (1), and a small myeloblast<br />

(2) in chronic myelomonocytic leukemia (CMML, see p. 107).<br />

89<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

c


90<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Acute Leukemias<br />

Acute leukemias are described in this place for morphological reasons, because<br />

they involve a predominance <strong>of</strong> mononuclear cells (p. 63). Although—or<br />

perhaps because—the term “leukemia” is relatively imprecise,<br />

an overview seems required (Table 13).<br />

The cellular phenomenon common to the different forms <strong>of</strong> leukemia is<br />

the rapidly progressive reduction in numbers <strong>of</strong> mature granulocytes,<br />

thrombocytes, and erythrocytes. Simultaneously, the leukocyte count<br />

usually increases due to the occurrence <strong>of</strong> atypical round cells.<br />

Table 13 Overview <strong>of</strong> all forms <strong>of</strong> leukemia<br />

Type <strong>of</strong> leukemia Classification/clinical findings<br />

Acute leukemias (ALAML, ALL)<br />

– Myeloid M0-7 (incl. monocytic, erythroid,<br />

megakaryoblastic leukemias)<br />

Always acute disease, <strong>of</strong>ten with<br />

fever and tendency to hemorrhage<br />

– Lymphocytic There may be lymphoma and thymus<br />

infiltrates<br />

Chronic myeloid leukemia (CML)<br />

– Persistent leukemic diseases <strong>of</strong> the<br />

myeloproliferative system (p. 114)<br />

Chronic lymphocytic leukemia (CLL)<br />

and other leukemic lymphomas<br />

– B-CLL (90%), T-CLL (p.74 f.)<br />

– Prolymphocytic leukemia (p.77)<br />

– Hairy cell leukemia (p.80)<br />

Chronic myelomonocytic leukemia<br />

(CMML)<br />

– Leukemic form <strong>of</strong> myelodysplasia<br />

(p.106) is classified between<br />

myelodysplasia and myeloproliferative<br />

diseases<br />

Chronic disease, usually with<br />

splenomegaly<br />

– Chronic lymphocytic leukemia,<br />

(rarely) prolymphocytic<br />

leukemia and hairy cell leukemia<br />

are primary leukemic lymphomas<br />

with a chronic course.<br />

All other non-Hodgkin lymphomas<br />

can develop secondary<br />

leukemic disease<br />

Subacute disease with transitions<br />

into acute forms <strong>of</strong> myeloid<br />

leukemia (secondary AML following<br />

MDS)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Predominance <strong>of</strong> Mononuclear Round to Oval Cells<br />

Note that in about one-fourth <strong>of</strong> all leukemias total leukocyte counts are<br />

normal, or even reduced, and the atypical round cells affect only the relative<br />

(differential) blood analysis (“aleukemic leukemia”).<br />

In all forms <strong>of</strong> leukemia, the more fluffy layered areas <strong>of</strong> a smear show that<br />

the nuclear chromatin structure is not dense and coarse, as in a normal<br />

lymphocyte nucleus (p. 49), but more delicately structured and irregular,<br />

<strong>of</strong>ten “sand-like”. A careful blood cell analysis should be carried out—perhaps<br />

with the assistance <strong>of</strong> a specialist laboratory—before bone marrow<br />

analysis is performed.<br />

In most cases, the high leukocyte count facilitates the diagnosis <strong>of</strong><br />

leukemia. Apart from the leukemia-specific blast cells, a variable number<br />

<strong>of</strong> segmented neutrophilic granulocytes may also remain, depending on<br />

the disease progression at the time <strong>of</strong> diagnosis. This gap in the cell series<br />

between blasts and mature cells is called “leukemic hiatus.” It is found in<br />

ALL but not in reactive responses or chronic myeloid leukemia, which<br />

show a continuous left shift. Morphological or differential diagnosis <strong>of</strong><br />

acute leukemia is followed by the diagnostic work-up that continues with<br />

cytochemical tests. Immunological identification <strong>of</strong> leukemia cells is always<br />

indicated (Table 14).<br />

Diagnostic work-up when acute leukemia is suspected:<br />

CBC, cytochemistry, bone marrow. Collection <strong>of</strong> material to identify cell<br />

surface markers, cytogenetics, molecular genetics.<br />

Morphological and Cytochemical Cell Identification<br />

After a first-line diagnosis <strong>of</strong> acute leukemia has been arrived at on the<br />

basis <strong>of</strong> the cell morphology (see above), the diagnosis must be refined by<br />

cytochemical testing <strong>of</strong> blood cells or bone marrow (on fresh smears).<br />

Table 14 shows a leukemia classification based on both morphological and<br />

cytochemical criteria. The table shows that a peroxidase test allows<br />

leukemias to be classified as peroxidase-positive (myeloid or monocytic)<br />

or peroxidase-negative (lymphoblastic). The next step is the immunological<br />

classification based on cell markers.<br />

In routine clinical hematology, the FAB classification (Table 14) will be<br />

with us for a few more years.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

91


92<br />

Table 14 Classification <strong>of</strong> the acute nonlymphatic leukemias by morphology, cytochemistry, and<br />

immunology<br />

FAB type* Peroxidase PAS α-Naphthylacetatesterase<br />

M0<br />

M1<br />

M2<br />

M3<br />

M4<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

AML with minimal<br />

marker differentiation,undifferentiated<br />

blasts<br />

without granules;<br />

distinguished from<br />

M1 and ALL only by<br />

immunophenotyping<br />

AML with distinct<br />

marker differentiation<br />

(but without morphologicaldifferentiation);<br />

sporadic discrete<br />

cytoplasmic<br />

granulation possible<br />

AML with morphologically<br />

mature<br />

cells; 10% <strong>of</strong> the<br />

blasts contain very<br />

small granules<br />

Acute promyelocytic<br />

leukemia; the<br />

predominant promyelocytes<br />

contain<br />

copious granules,<br />

some contain Auer<br />

bodies; variant M3<br />

contains few<br />

granules; peripheral<br />

bilobal blasts<br />

Acute myelomonocytic<br />

leukemia;<br />

30–80% <strong>of</strong> bone<br />

marrow blasts are<br />

myeloblasts, promyelocytes,<br />

and<br />

myelocytes; 20%<br />

are monocytes; variant<br />

M4 eosinophilia;<br />

additional immature<br />

eosinophils<br />

with dark granules<br />

Naphthyl-ASDesterase<br />

Immunophenotype<br />

3% CD 13 or<br />

CD 33 or<br />

MPO <br />

CD 79 a <br />

and cyCD 3 <br />

and cyCD 22 <br />

and CD 61/<br />

CD 41 and<br />

CD 14 <br />

3% Negative<br />

to fine<br />

gran.<br />

3% Negative<br />

to fine<br />

gran.<br />

MPO and<br />

CD 13/CD 33/<br />

CD 65s /<br />

and CD 14 <br />

MPO and<br />

CD 13/CD 33/<br />

CD 65s/<br />

CD 15 /<br />

and CD 14 <br />

100% MPO and<br />

CD 13 and<br />

CD 33 and<br />

normally CD<br />

34 and<br />

HLA-DR <br />

3% +<br />

20%<br />

+ Mixed M 1/<br />

M 2 and M 5<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Table 14 Continued<br />

FAB type* Peroxidase PAS α-Naphthylacetateesterase<br />

M5 a) Acute monoblasticleukemia;<br />

monoblasts<br />

predominant in<br />

the blood and<br />

bone marrow<br />

b) Acute monocytic<br />

leukemia;<br />

monocytes in<br />

the process <strong>of</strong><br />

maturation predominante<br />

M6<br />

M7<br />

Acute erythroid<br />

leukemia; 50% <strong>of</strong><br />

bone marrow blasts<br />

are erythropoietic,<br />

30% myeloblasts<br />

Acute megakaryocytic<br />

leukemia; very<br />

polymorphic, sometimes<br />

vacuolated<br />

blasts, some with<br />

cytoplastic blebs,<br />

sometimes aggregated<br />

with thrombocytes<br />

Predominance <strong>of</strong> Mononuclear Round to Oval Cells<br />

+++<br />

80%<br />

+++ +++<br />

Naphthyl-ASDesterase<br />

Immunophenotype<br />

93<br />

+++ CD 13/CD 33/<br />

CD 65/CD 14/<br />

CD64 /<br />

and HLA-DR<br />

<br />

Erythroblasts<br />

Gly A and<br />

CD 36 <br />

Myeloblasts<br />

MPO/CD<br />

13/CD 33/CD<br />

65s / and<br />

CD 14 <br />

CD 13/CD 33<br />

/ und CD<br />

41 oder CD<br />

61 <br />

* French American British Classification (FAB) 1976/85<br />

** A biphenotypic leukemia must be considered a possibility if several additional lymphoblastic<br />

markers are present<br />

PAS Periodic acid-Schiff reaction<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


94<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Chromosome analysis provides important information. In practice,<br />

however, the diagnosis <strong>of</strong> acute leukemia is still based on morphological<br />

criteria.<br />

However, where the possibilities <strong>of</strong> modern therapeutic and prognostic<br />

methods are fully accessible, new laboratory procedures based on genetic<br />

and molecular biological testing procedures form part <strong>of</strong> the diagnostic<br />

work-up <strong>of</strong> AML. The current WHO classification takes account <strong>of</strong> these<br />

new methods, placing genetic, morphological, and anamnestic findings in<br />

a hierarchical order (Table 15).<br />

According to the new WHO classification, blasts account for more than<br />

20% <strong>of</strong> cells in acute myeloid leukemia (in contradistinction to myelodysplasias).<br />

Table 15 WHO classification <strong>of</strong> AML<br />

AML with specific<br />

cytogenetic translocations<br />

AML with dysplasia in<br />

more than 1 cell line<br />

(2 or 3 cell lines<br />

affected)*<br />

Therapy-induced AML<br />

und MDS<br />

AML that does not fit<br />

any <strong>of</strong> the other categories<br />

– With t(8;21) (q22; q22), AML 1/ETO<br />

– Acute promyelocytic leukemia (AML M3 with t(15;17) (q22;<br />

q11-12) and variants, PML/RAR-α<br />

– With abnormal bone marrow eosinophils and (inv16)<br />

(p13;q22) or to t(16;16) (p13; q22); CBF/MYH 11<br />

– With 11q23 (MLL) anomalies<br />

– With preceding myelodysplastic/myeloproliferative syndrome<br />

– Without preceding myelodysplastic syndrome<br />

– After treatment with alkylating agents<br />

– After treatment with epipodophyllotoxin<br />

– Other triggers<br />

– AML, minimal differentiation<br />

– AML without cell maturation<br />

– AML with cell maturation<br />

– Acute myelomonocytic leukemia<br />

– Acute monocytic leukemia<br />

– Acute erythroid leukemia<br />

– Acute megakaryoblastic leukemia<br />

– Acute panmyelosis with myel<strong>of</strong>ibrosis<br />

– Myelosarcoma/chloroma<br />

– Acute biphenotypic leukemia**<br />

* The dysplasia must be evident in at least 50% <strong>of</strong> the bone marrow cells and in 2–3 cell lines.<br />

** Biphenotypic leukemias should be classified according to their immunophenotypes. They<br />

are grouped between acute lymphocytic and acute myeloid leukemias.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Predominance <strong>of</strong> Mononuclear Round to Oval Cells<br />

Acute Myeloid Leukemias (AML)<br />

95<br />

Morphological analysis makes it possible to group the predominant<br />

leukemic cells into myeloblasts and promyeloblasts, monocytes, or atypical<br />

(lympho)blasts. A morphological subclassification <strong>of</strong> these main<br />

groups was put forward in the French–American–British (FAB) classification<br />

(Table 14).<br />

In practical, treatment-oriented terms, the most relevant factor is<br />

whether the acute leukemia is characterized as myeloid or lymphatic.<br />

Including the very rare forms, there are at least 11 forms <strong>of</strong> myeloid<br />

leukemia.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


96 Abnormalities <strong>of</strong> the White Cell Series<br />

Acute Myeloblastic Leukemia (Type M0 through M2 in the FAB Classification).<br />

Morphologically, the cell populations that dominate the CBC and<br />

bone marrow analyses (Fig. 31) more or less resemble myeloblasts in the<br />

course <strong>of</strong> normal granulopoiesis. Differences may be found to varying<br />

degrees in the form <strong>of</strong> coarser chromatin structure, more prominently defined<br />

nucleoli, and relatively narrow cytoplasm. Compared with lymphocytes<br />

(micromyeloblasts), the analyzed cells may be up to threefold larger.<br />

In a good smear, the transformed cells can be distinguished from lymphatic<br />

cells by their usually reticular chromatin structure and its irregular<br />

organization. Occasionally, the cytoplasm contains crystalloid azurophilic<br />

needle-shaped primary granules (Auer bodies). Auer bodies (rods) are<br />

conglomerates <strong>of</strong> azurophilic granules. A few cells may begin to display<br />

promyelocytic granulation. Cytochemistry shows that from stage M1 onward,<br />

more than 3% <strong>of</strong> the blasts are peroxidase-positive.<br />

Characteristics <strong>of</strong> Acute Leukemias<br />

Age <strong>of</strong> onset: Any age.<br />

Clinical findings: Fatigue, fever, and signs <strong>of</strong> hemorrhage in later<br />

stages.<br />

Lymph node and mediastinal tumors are typical only in ALL.<br />

Generalized involvement <strong>of</strong> all organs (sometimes including the<br />

meninges) is always present.<br />

CBC and laboratory: Hb , thrombocytes , leukocytes usually<br />

strongly elevated (~ 80%) but sometimes decreased or normal.<br />

In the differential blood analysis, blasts predominate (morphologies<br />

vary).<br />

Beware: Extensive urate accumulation!<br />

Further diagnostics: Bone marrow, cytochemistry, immunocytochemistry,<br />

cytogenetics, and molecular genetics.<br />

Differential diagnosis: Transformed myeloproliferative syndrome<br />

(e.g., CML) or myelodysplastic syndrome.<br />

Leukemic non-Hodgkin lymphomas (incl. CLL).<br />

Aplastic anemias.<br />

Tumors in the bone marrow (carcinomas, but also rhabdomyosarcoma).<br />

Course, therapy: Usually rapid progression with infectious complications<br />

and bleeding.<br />

Immediate efficient chemotherapy in a hematology facility; bone<br />

marrow transplant may be considered, with curative intent.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

Fundamental characteristic <strong>of</strong> acute leukemia: variable blasts<br />

drive out other cell series<br />

c<br />

d<br />

Fig. 31 Acute leukemia, M0–M2. a Undifferentiated blast with dense, fine chromatin,<br />

nucleolus (arrow), and narrow basophilic cytoplasm without granules. This<br />

cell type is typical <strong>of</strong> early myeloid leukemia (M0–M1); the final classification is<br />

made using cell surface marker analysis (see Table 14). b The peroxidase reaction,<br />

characteristic <strong>of</strong> cells in the myeloid series, shows positive ( 3%) only for stage<br />

M1 leukemia and higher. The image shows a weakly positive blast (1), strongly positive<br />

eosinophil (2), and positive myelocyte (3). c and d Variants <strong>of</strong> M2 leukemia.<br />

Some <strong>of</strong> the cells already contain granules (1) and crystal-like Auer bodies (2).<br />

97<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

b


98 Abnormalities <strong>of</strong> the White Cell Series<br />

Acute Promyelocytic Leukemia (FAB Classification Type M3 and M3 v). The<br />

characteristic feature <strong>of</strong> the cells, which are usually quite large with variably<br />

structured nuclei, is extensive promyelocytic granulation. Auer rods<br />

are commonly present. Cytochemistry reveals a positive peroxidase reaction<br />

for almost all cells. All other reactions are nonspecific. Acute leukemia<br />

with predominantly bilobed nuclei is classified as a variant <strong>of</strong> M3 (M3 V).<br />

The cytoplasm may appear either ungranulated (M3) or very strongly<br />

granulated (M3 V).<br />

Acute Myelomonocytic Leukemia (FAB Classification Type M4). Given the<br />

close relationship between cells in the granulopoietic and the monocytopoietic<br />

series (see p. 3), it would not be surprising if the these two systems<br />

showed a common alteration in leukemic transformation. Thus, acute myelomonocytic<br />

leukemia shows increased granulocytopoiesis (up to more<br />

than 20% myeloblasts) with altered cell morphologies, together with increased<br />

monocytopoiesis yielding more than 20% monoblasts or promonocytes.<br />

Immature myeloid cells (atypical myelocytes to myeloblasts)<br />

are found in peripheral blood in addition to monocyte-related cells. Cytochemically,<br />

the classification calls for more than 3% peroxidase-positive<br />

and more than 20% esterase-positive blasts in the bone marrow. M4 is similar<br />

to M2; the difference is that in the M4 type the monocyte series is<br />

strongly affected. In addition to the above characteristics, the M4Eo variant<br />

shows abnormal eosinophils with dark purple staining granules.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

b<br />

The diagnosis <strong>of</strong> acute leukemia is relevant even without further<br />

subclassification<br />

d<br />

f<br />

Fig. 32 Acute leukemia M3 and M4. a Blood analysis in promyelocytic leukemia<br />

(M3): copious cytoplasmic granules. b In type M3, multiple Auer bodies are <strong>of</strong>ten<br />

stacked like firewood (so-called faggot cells). c Blood analysis in variant M3 v with<br />

dumbbell-shaped nuclei. Auer bodies d Bone marrow cytology in acute myelomonocytic<br />

leukemia M4: in addition to myeloblasts (1) and promyelocytes (2) there<br />

are also monocytoid cells (3). e In variant M4Eo abnormal precursors <strong>of</strong> eosinophils<br />

with dark granules are present. f Esterase as a marker enzyme for the monocyte<br />

series in M4 leukemia.<br />

99<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

c<br />

e


100<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Acute Monocytic Leukemia (FAB Classification Types M5 a+b). Two morphologically<br />

distinct forms <strong>of</strong> acute monocytic leukemias exist, monoblastic<br />

and monocytic. In the monoblastic variant M5a, blasts predominate in the<br />

blood and bone marrow. The blast nuclei show a delicate chromatin structure<br />

with several nucleoli. Often, only the faintly grayish-blue stained cytoplasm<br />

hints at their derivation.<br />

In monocytic leukemia (type M5b), the bone marrow contains promonocytes,<br />

which are similar to the blasts in monocytic leukemia, but their nuclei<br />

are polymorphic and show ridges and lobes. Some promonocytes<br />

show faintly stained azurophilic granules. The peripheral blood contains<br />

monocytoid cells in different stages <strong>of</strong> maturation which cannot be distinguished<br />

with certainty from normal monocytes. Both types are characterized<br />

by strong positive esterase reactions in over 80% <strong>of</strong> the blasts,<br />

whereas the peroxidase reactivity is usually negative, or positive in only a<br />

few cells.<br />

Acute Erythroleukemia (FAB Classification Type M6)<br />

Erythroleukemia is a malignant disorder <strong>of</strong> both cell series. It is suspected<br />

when mature granulocytes are virtually absent, but blasts (myeloblasts)<br />

are present in addition to nucleated erythrocyte precursors, usually<br />

erythroblasts (for morphology, see p. 33). The bone marrow is completely<br />

overwhelmed by myeloblasts and erythroblasts (more than 50% <strong>of</strong> cells in<br />

the process <strong>of</strong> erythropoiesis). Bone marrow cytology and cytochemistry<br />

confirm the diagnosis. Sporadically, some cases show granulopenia,<br />

erythroblasts, and severely dedifferentiated blasts, which correspond to<br />

immature red cell precursors (proerythroblasts and macroblasts).<br />

The differential diagnosis in cases <strong>of</strong> cytopenia with red blood cell precursors<br />

found in the CBC must include bone marrow carcinosis, in which<br />

the bone marrow–blood barrier is destroyed and immature red cells (and<br />

sometimes white cells) appear in the bloodstream. Bone marrow cytology<br />

and/or bone marrow histology clarifies the diagnosis. Hemolysis with hypersplenism<br />

can also show this constellation <strong>of</strong> signs.<br />

Fig. 33 Acute leukemia M5 and M6. a In monoblastic leukemia M5 a, blasts with a<br />

fine nuclear structure and wide cytoplasm dominate the CBC. b Seemingly mature<br />

monocytes in monocytic leukemia M5 b. c Homogeneous infiltration <strong>of</strong> the<br />

bone marrow by monoblasts (M5 a). Only residual granulopoiesis (arrow). d Same<br />

as c but after esterase staining. The stage M5 a blasts show a clear positive reaction<br />

(red stain). There is a nonspecific-esterase (NSE)-negative promyelocyte.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

b<br />

d<br />

e<br />

Acute leukemias may also derive from monoblasts or erythroblasts<br />

Fig. 33 e Same as c Only the myelocyte in the center stains peroxidase-positive<br />

(brown tint); the monoblasts are peroxidase-negative. f In acute erythrocytic leukemia<br />

(M6) erythroblasts and myeloblasts are usually found in the blood. This<br />

image <strong>of</strong> bone marrow cytology in M6 shows increased, dysplastic erythropoiesis<br />

(e.g., 1) in addition to myeloblasts (2).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

101<br />

c<br />

f


102<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Acute Megakaryoblastic Leukemia (FAB Classification Type M7)<br />

This form <strong>of</strong> leukemia is very rare in adults and occurs more <strong>of</strong>ten in<br />

children. It can also occur as “acute myel<strong>of</strong>ibrosis,” with rapid onset <strong>of</strong><br />

tricytopenia and usually small-scale immigration into the blood <strong>of</strong> dedifferentiated<br />

medium-sized blasts without granules. Bone marrow<br />

harvesting is difficult because the bone marrow is very fibrous. Only bone<br />

marrow histology and marker analysis (fluorescence-activated cell<br />

sorting, FACS) can confirm the suspected diagnosis.<br />

The differential diagnosis, especially if the spleen is very enlarged,<br />

should include the megakaryoblastic transformation <strong>of</strong> CML or osteomyelosclerosis<br />

(see pp. 112 ff.), in which blast morphology is very similar.<br />

AML with Dysplasia<br />

The WHO classification (p. 94) gives a special place to AML with dysplasia<br />

in two to three cell series, either as primary syndrome or following a myelodysplastic<br />

syndrome (see pp. 106) or a myeloproliferative disease (see<br />

pp. 114 ff.).<br />

Criteria for dysgranulopoiesis: 50% <strong>of</strong> all segmented neutrophils<br />

have no granules or very few granules, or show the Pelger anomaly, or are<br />

peroxidase-negative.<br />

Criteria for dyserythropoiesis: 50% <strong>of</strong> the red cell precursor cells<br />

display one <strong>of</strong> the following anomalies: karyorrhexis, megaloblastoid<br />

traits, more than one nucleus, nuclear fragmentation.<br />

Criteria for dysmegakaryopoiesis: 50% <strong>of</strong> at least six megakaryocytes<br />

show one <strong>of</strong> the following anomalies: micromegakaryocytes, more<br />

than one separate nucleus, large mononuclear cells.<br />

Hypoplastic AML<br />

Sometimes (mostly in the mild or “aleukemic” leukemias <strong>of</strong> the FAB or<br />

WHO classifications), the bone marrow is largely empty and shows only a<br />

few blasts, which usually occur in clusters. In such a case, a very detailed<br />

analysis is essential for a differential diagnosis versus aplastic anemia (see<br />

pp. 148 f.).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

New WHO classification: AML with dysplasia and hypoplastic<br />

AML<br />

Fig. 34 AML with dysplasia and hypoplastic AML. a AML with dysplasia: megaloblastoid<br />

(dysplastic) erythropoiesis (1) and dysplastic granulopoiesis with Pelger-<br />

Huët forms (2) and absence <strong>of</strong> granulation in a myelocyte (3). Myeloblast (4).<br />

b Multiple separated nuclei in a megakaryocyte (1) in AML with dysplasia. Dyserythropoiesis<br />

with karyorrhexis (2). c and d Hypoplastic AML. c Cell numbers below<br />

normal for age in the bone marrow. d Magnification <strong>of</strong> the area indicated in c,<br />

showing predominance <strong>of</strong> undifferentiated blasts (e.g., 1).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

103<br />

b<br />

d


104<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Acute Lymphoblastic Leukemia (ALL)<br />

ALL are the leukemias in which the cells do not morphologically resemble<br />

myeloblasts, promyelocytes, or monocytes, nor do they show the corresponding<br />

cytochemical pattern. Common attributes are a usually slightly<br />

smaller cell nucleus and denser chromatin structure, the grainy consistency<br />

<strong>of</strong> which can be made out only with optimal smear technique (i.e.,<br />

very light). The classification as ALL is based on the (<strong>of</strong>ten remote) similarities<br />

<strong>of</strong> the cells to lymphocytes or lymphoblasts from lymph nodes, and<br />

on their immunological cell marker behavior. Insufficiently close morphological<br />

analysis can also result in possible confusion with chronic lymphocytic<br />

leukemia (CLL), but cell surface marker analysis (see below) will correct<br />

this mistake. Advanced diagnostics start with peroxidase and esterase<br />

tests on fresh smears, performed in a hematology laboratory, together<br />

with (as a minimum) immunological marker studies carried out on fresh<br />

heparinized blood samples in a specialist laboratory. The detailed differentiation<br />

provided by this cell surface marker analysis has prognostic implications<br />

and some therapeutic relevance especially for the distinction to<br />

bilineage leukemia and AML (Table 16).<br />

Table 16 Immunological classification <strong>of</strong> acute bilineage leukemias (adapted from Bene MC<br />

et al. (1995) European Group for the Immunological Characterization <strong>of</strong> Leukemias (EGIL) 9:<br />

1783–1786)<br />

Score B-lymphoid T-lymphoid Myeloid<br />

2 CytCD79a* CD3(m/cyt) MP0<br />

Cyt IgM anti-TCR<br />

1 CD19 CD2 CD117<br />

CD20 CD5 CD13<br />

CD10 CD8 CD33<br />

CD10 CD65<br />

0.5 TdT TdT CD14<br />

CD24 CD7 CD15<br />

CD1a CD64<br />

* CD79a may also be expressed in some cases <strong>of</strong> precursor T-lymphoblastic leukemia/lymphoma.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

The cells in acute lymphocytic leukemia vary, and the subtypes<br />

can be reliably identified only by immunological methods<br />

c<br />

d<br />

Fig. 35 Acute lymphocytic leukemias. a Screening view: blasts (1) and lymphocytes<br />

(2) in ALL. Further classification <strong>of</strong> the blasts requires immunological methods<br />

(common ALL). b Same case as a . The blasts show a dense, irregular nuclear<br />

structure and narrow cytoplasm (cf. mononucleosis, p. 69). Lymphocyte (2). c ALL<br />

blasts with indentations must be distinguished from small-cell non-Hodgkin<br />

lymphoma (e.g., mantle cell lymphoma, p. 77) by cell surface marker analysis.<br />

d Bone marrow: large, vacuolated blasts, typical <strong>of</strong> B-cell ALL. The image shows<br />

residual dysplastic erythropoietic cells (arrow).<br />

105<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

b


106<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Myelodysplasia (MDS)<br />

Clinical practice has long been familiar with the scenario in which, after<br />

years <strong>of</strong> bone marrow insufficiency with a more or less pronounced deficit<br />

in all three cell series (tricytopenia), patients pass into a phase <strong>of</strong> insidiously<br />

increasing blast counts and from there into frank leukosis<br />

—although the evolution may come to a halt at any <strong>of</strong> these stages. The<br />

transitions between the forms <strong>of</strong> myelodysplastic syndromes are very<br />

fluid, and they have the following features in common:<br />

➤ Anemia, bicytopenia, or tricytopenia without known cause.<br />

➤ Dyserythropoiesis with sometimes pronounced erythrocyte anisocytosis;<br />

in the bone marrow <strong>of</strong>ten megaloblastoid cells and/or ring sideroblasts.<br />

➤ Dysgranulopoiesis with pseudo-Pelger-Huët nuclear anomaly (hyposegmentation)<br />

and hypogranulation (<strong>of</strong>ten no peroxidase reactivity) <strong>of</strong><br />

segmented and band granulocytes in blood and bone marrow.<br />

➤ Dysmegakaryopoiesis with micromegakaryocytes.<br />

The FAB classification is the best-known scheme so far for organizing the<br />

different forms <strong>of</strong> myelodysplasia (Table 17).<br />

Table 17 Forms <strong>of</strong> myelodysplasia<br />

Form <strong>of</strong> myelodysplasia Blood analysis Bone marrow<br />

RA = refractory anemia Anemia (normochromic<br />

or hyperchromic);<br />

possibly<br />

pseudo-Pelger granulocytes;<br />

blasts 1%<br />

RAS = refractory anemia<br />

with ring sideroblasts<br />

( aquired<br />

idiopathic sideroblastic<br />

anemia, p. 137)<br />

RAEB = refractory anemia<br />

with excess <strong>of</strong><br />

blasts<br />

Hypochromic and<br />

hyperchromic erythrocytes<br />

side by side,<br />

sometimes discrete<br />

thrombopenia and<br />

leukopenia; pseudo-<br />

Pelger cells<br />

Often thrombocytopenia<br />

in addition to<br />

anemia; blasts 5%,<br />

monocytes 1000/µl,<br />

pseudo-Pelger syndrome<br />

Dyserythropoiesis<br />

(marginal dysgranulopoiesis<br />

and dysmegakaryopoiesis<br />

10%)<br />

5% blasts<br />

More than 15% <strong>of</strong> the<br />

red cell precursors are<br />

ring sideroblasts;<br />

blasts 5%<br />

Erythropoietic hyperplasia<br />

(with or without<br />

ring sideroblasts);<br />

5–20% blasts<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

Cont. p. 108


a<br />

c<br />

In unexplained anemia and/or leukocytopenia and/or thrombocytopenia,<br />

blood cell abnormalities may indicate myelodysplasia<br />

Fig. 36 Myelodysplasia and CMML. a–d Different degrees <strong>of</strong> abnormal maturation<br />

(pseudo-Pelger type); the nuclear density can reach that <strong>of</strong> erythroblasts<br />

(d). The cytoplasmic hypogranulation is also observed in normal segmented granulocytes.<br />

These abnormalities are seen in myelodysplasia or after chemotherapy,<br />

among other conditions. e Blood analysis in CMML: monocytes (1), promyelocyte<br />

(2), and pseudo-Pelger cell (3). Thrombocytopenia.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

107<br />

b<br />

d<br />

e


108<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Table 17 Continued<br />

Form <strong>of</strong> myelodysplasia Blood analysis Bone marrow<br />

CMML = chronic myelomonocytic<br />

leukemia<br />

RAEB in transformation<br />

(RAEBt)*<br />

Blasts 5%, monocytes<br />

1000/µl,<br />

pseudo-Pelger syndrome<br />

Similar to RAEB but<br />

5% blasts<br />

Hypercellular, blasts<br />

20%, elevated promonocytes<br />

Blasts 20–30% (some<br />

cells contain Auer<br />

bodies)<br />

* In the WHO classification, the category RAEBt would belong to the category <strong>of</strong> acute myeloid<br />

leukemia.<br />

The new WHO classification <strong>of</strong> myelodysplastic syndromes defines the<br />

differences in cell morphology even more precisely than the FAB classification<br />

(Table 18).<br />

For the criteria <strong>of</strong> dysplasia, see page 106.<br />

The “5q- syndrome” is highlighted as a specific type <strong>of</strong> myelodysplasia<br />

in the WHO classification; in the FAB classification it would be a subtype <strong>of</strong><br />

RA and RAS. A macrocytic anemia, the 5q- syndrome manifests with normal<br />

or increased thrombocyte counts while the bone marrow contains<br />

megakaryocytes with hyposegmented round nuclei (Fig. 37 b).<br />

Naturally, bone marrow analysis is <strong>of</strong> particular importance in the myelodysplasias.<br />

Table 18 WHO classification <strong>of</strong> myelodysplastic syndromes<br />

Disease* Dysplasia** Blasts in<br />

peripheral<br />

blood<br />

Blasts in the<br />

bone marrow<br />

Ring sideroblasts<br />

in the<br />

bone marrow<br />

Cytogenetics<br />

5q- syndrome Usually only E 5% 5% 15% 5q only<br />

RA Usually only DysE 1% 5% 15% Variable<br />

RARS Usually only DysE None 5% 15% Variable<br />

RCMD 2–3 lines Rarely 5% 15% Variable<br />

RCMD-RS 2–3 lines Rarely 5% 15% Variable<br />

RAEB-1 1–3 lines 5% 5–9% 15% Variable<br />

RAEB-2 1–3 lines 5–19% 10–19% 15% Variable<br />

CMML-1 1–3 lines 5% 10% 15% Variable<br />

CMML-2 1–3 lines 5–19% 10–19% 15% Variable<br />

MDS-U 1 cell lineage None 5% 15% Variable<br />

* RA = refractory anemia; RARS = refractory anemia with ring sideroblasts; RCMD = refractory<br />

cytopenia with more than one dysplastic cell line; RCMD-RC = refractory cytopenia with more<br />

than one dysplastic lineage and ring sideroblasts; RAEB = refractory anemia with elevated blast<br />

count; CMML = chronic myelomonocytic leukemia, persistent monocytosis (more than<br />

1 10 9 /l) in peripheral blood; MDS-U = MDS, unclassifiable. ** Dysplasia in granulopoiesis<br />

= Dys G, in erythropoiesis = DysE, in megakaryopoiesis = DysM, multilineage dysplasia<br />

= two cell lines affected; trilineage dysplasia (TLD) = all three lineage show dysplasia.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

The classification <strong>of</strong> myelodysplasias requires bone marrow analysis<br />

c<br />

d<br />

Fig. 37 Bone marrow analysis in myelodysplasia. a Dysmegakaryopoiesis in<br />

myelodysplastic syndrome (MDS). Relatively small disk-forming megakaryocytes<br />

(1) and multiple singular nuclei (2) are <strong>of</strong>ten seen. b Mononuclear megakaryocytes<br />

(frequent in 5q-syndrome). c Dyserythropoiesis. Particularly striking is the<br />

coarse nuclear structure with very light gaps in the chromatin (arrow 1). Some are<br />

megaloblast-like but coarser (arrow 2). d Iron staining <strong>of</strong> the bone marrow (Prussian<br />

blue) in myelodysplasia <strong>of</strong> the RARS type: dense iron granules forming a partial<br />

ring around the nuclei (ring sideroblasts).<br />

109<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

b


110<br />

Table 19 Diagnostic work-up for anomalies in the white cell series with polynucleated<br />

(segmented) cells predominating<br />

Clinical findings Hb MCH Leukocytes<br />

Acute temperature,<br />

possibly<br />

focal signs<br />

Patient smokes<br />

heavily (no<br />

splenomegaly)<br />

Slowly developing<br />

fatigue,<br />

spleen <br />

Slowly developing<br />

fatigue,<br />

spleen <br />

Segmented<br />

cells<br />

(%)<br />

Lymphocytes<br />

(%)<br />

Other cells<br />

n n Left shift<br />

n/ n <br />

/n /n Left shift<br />

n// n// Normoblasts,<br />

left shift<br />

n Some normoblasts<br />

Pruritus or<br />

exanthema<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Prevalence <strong>of</strong> Polynuclear<br />

(Segmented) Cells (Table 19)<br />

Neutrophilia without Left Shift<br />

For clarity, conditions in which mononuclear cells (lymphocytes, monocytes)<br />

predominate were in the previous section kept distinct from hematological<br />

conditions in which cells with segmented nuclei and, in some<br />

cases, their precursors predominate. Leukocytosis with a predominance<br />

<strong>of</strong> segmented neutrophilic granulocytes without the less mature forms is<br />

called granulocytosis or neutrophilia.<br />

n n n/ n n <br />

Eosinophils<br />

Diagnostic steps proceed from left to right. ⎪ The next step is usually unnecessary; the next step<br />

is obligatory.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Causes <strong>of</strong> Neutrophilia<br />

➤ All kinds <strong>of</strong> stress<br />

➤ Pregnancy<br />

➤ Connective tissue diseases<br />

➤ Tissue necrosis, e.g., after myocardial or pulmonary infarction<br />

➤ Acidosis <strong>of</strong> various etiologies, e.g., nephrogenic diabetes insipidus<br />

➤ Medications, drugs, or noxious chemicals, such as<br />

– Nicotine<br />

– Corticosteroids<br />

– Adrenaline<br />

– Digitalis<br />

– Allopurinol<br />

Thrombocytes<br />

Electrophoresis<br />

Prevalence <strong>of</strong> Polynuclear (Segmented) Cells<br />

Tentative<br />

diagnosis<br />

n n Acute bacterial<br />

infection<br />

(possibly with<br />

leukemoid<br />

reaction)<br />

n/ n Smoker’s<br />

leukocytosis<br />

n// n Chronic<br />

myeloid<br />

leukemia<br />

(CML)<br />

n// n Osteomyelosclerosis<br />

n/ n Polycythemia<br />

with concomitantleukocytosis<br />

n n Reactive<br />

eosinophilia<br />

– Barbiturates<br />

– Lithium<br />

– Streptomycin<br />

– Sulfonamide<br />

Evidence/advanced<br />

diagnostics<br />

Search for disease<br />

focus<br />

Bone<br />

marrow<br />

Anamnesis In progressive<br />

disease: bone<br />

marrow analysis<br />

(DD myeloproliferative<br />

disease)<br />

Cytogenetics,<br />

BCR-ABL<br />

Tear-drop-shaped<br />

erythrocytes<br />

Search for disease<br />

focus/allergen<br />

Complete<br />

bone marrow<br />

analysis, all<br />

fractions<br />

Often dry tap<br />

bone marrow<br />

histology<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

Ref.<br />

page<br />

p. 112<br />

p. 112<br />

p. 116<br />

p. 122<br />

p. 162<br />

p. 124<br />

111


112<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Reactive Left Shift<br />

A relative left shift in the granulocyte series means less mature forms in<br />

excess <strong>of</strong> 5% band neutrophils; the preceding, less differentiated cell forms<br />

are included and all transitional forms are taken into account. This left<br />

shift almost always indicates an increase in new cell production in this cell<br />

series. In most cases, it is associated with a raised total leukocyte count.<br />

However, since total leukocyte counts are subject to various interfering<br />

factors that can also alter the cell distribution, left shift without leukocytosis<br />

can occur, and has no further diagnostic value. At best, if no other explanations<br />

<strong>of</strong>fer, a left shift without leukemia can prompt investigation for<br />

splenomegaly, which would have prevented elevation <strong>of</strong> the leukocyte<br />

count by increased sequestration <strong>of</strong> leukocytes as in hypersplenism.<br />

In evaluating the magnitude <strong>of</strong> a left shift, the basic principle is that the<br />

more immature the cell forms, the more rarely they appear; and that there<br />

is a continuum starting from segmented granulocytes and sometimes<br />

reaching as far as myeloblasts. Accordingly, a moderate left shift <strong>of</strong> medium<br />

magnitude may include myelocytes and a severe left shift may go as<br />

far as a few promyelocytes and (very rarely) myeloblasts, all depending on<br />

how fulminant the triggering process is and how responsive the individual.<br />

The term “pathological left shift” (for a left shift that includes<br />

promyelocytes and myeloblasts) is inappropriate, because such observations<br />

can reflect a very active physiological reaction, perhaps following a<br />

“leukemoid reaction” with pronounced left shift and leukocytosis.<br />

Causes <strong>of</strong> Reactive Left Shift<br />

Left shift occurs regularly in the following situations:<br />

➤ Bacterial infections (including miliary tuberculosis),<br />

➤ Nonbacterial inflammation (e.g., colitis, pancreatitis, phlebitis, and<br />

connective tissue diseases)<br />

➤ Cell breakdown (e.g., burns, liver failure, hemolysis)<br />

Left shift sometimes occurs in the following situations:<br />

➤ Infection with fungi, mycoplasm, viruses, or parasites<br />

➤ Myocardial or pulmonary infarction<br />

➤ Metabolic changes (e.g., pregnancy, acidosis, hyperthyroidism)<br />

➤ Phases <strong>of</strong> compensation and recuperation (hemorrhages, hemolysis,<br />

or after medical or radiological immunosuppression).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Predominance <strong>of</strong> the granulocytic lineage with copious granulation<br />

and sporadic immature cells: usually a reactive left shift<br />

Fig. 38 Left shift. a and b Typical blood smear after bacterial infection: toxic granulation<br />

in a segmented granulocyte (1), monocyte with gray–blue cytoplasm<br />

(2), metamyelocyte (3), and myelocyte (4). c Blood analysis in sepsis: promyelocyte<br />

(1) and orthochromatic erythroblast (2). Thrombocytopenia. d and e Reactive<br />

left shift as far as promyelocytes (1). Particularly striking are the reddish granules<br />

in a band neutrophilic granulocyte (2).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

113<br />

b<br />

d<br />

e


114<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Chronic Myeloid Leukemia and Myeloproliferative<br />

Syndrome (Chronic Myeloproliferative Disorders,<br />

CMPD)<br />

The chronic myeloproliferative disorders (previously also called the myeloproliferative<br />

syndromes) include chronic myeloid leukemia (CML),<br />

osteomyelosclerosis (OMS), polycythemia vera (PV) and essential thrombocythemia<br />

(ET). Clearly, noxious agents <strong>of</strong> unknown etiology affect the<br />

progenitor cells at different stages <strong>of</strong> differentiation and trigger chronic<br />

malignant proliferation in the white cell series (CML), the red cell series<br />

(PV), and the thrombocyte series (ET). Sometimes, they lead to concomitant<br />

synthesis <strong>of</strong> fibers (OMS). Transitional forms and mixed forms exist<br />

particularly between PV, ET, and OMS.<br />

The chronic myeloproliferative disorders encompass chronic autonomous<br />

disorders <strong>of</strong> the bone marrow and the embryonic blood-generating<br />

organs (spleen and liver), which may involve one or several cell lines.<br />

The common attributes <strong>of</strong> these diseases are onset in middle age, development<br />

<strong>of</strong> splenomegaly, and slow disease progression (Table 20).<br />

In 95% <strong>of</strong> cases, CML shows a specific chromosome aberration<br />

(Philadelphia chromosome with a specific BCR-ABL translocation) and<br />

may make the transition into a blast crisis.<br />

PV and ET <strong>of</strong>ten show similar traits (high thrombocyte count or high<br />

Hb) and have a tendency to secondary bone marrow fibrosis. OMS is primarily<br />

characterized by fibrosis in bone marrow and splenomegaly (see<br />

p. 122).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


115<br />

Table 20 Clinical characteristics and differential diagnostic criteria in chronic<br />

myeloproliferative disease<br />

Splenomegaly<br />

Changes in<br />

the CBC for<br />

granulopoiesis<br />

and/or<br />

erythropoiesis<br />

Thrombocytes<br />

Bone marrow<br />

cytology<br />

Bone marrow<br />

histology<br />

Philadelphia<br />

chromosome<br />

Other chromosomal<br />

alterations<br />

Alkaline<br />

Leukocytephosphatase<br />

(ALP)<br />

Vitamin B12<br />

900 pg/ml<br />

CML<br />

(see p. 116)<br />

PV<br />

(see p. 162)<br />

OMS<br />

(see p. 122)<br />

+ No + No<br />

Leukocytosis<br />

with left shift,<br />

eosinophilia,<br />

basophilia !!<br />

Leukocytosis,<br />

hematocrit<br />

!!<br />

() ()<br />

Giant forms<br />

Very hypercellular,<br />

basophilia,<br />

megakaryocytes,<br />

and<br />

eosinophilia<br />

<br />

Prevalence <strong>of</strong> Polynuclear (Segmented) Cells<br />

Granulopoiesis<br />

, megakaryocytes<br />

<br />

Markedly<br />

hypercellular,<br />

erythropoiesis<br />

Number <strong>of</strong><br />

cells <br />

Tear-dropshapederythrocytes,<br />

left<br />

shift in the<br />

granulopoiesis,<br />

normoblasts<br />

ET<br />

(see p. 170)<br />

No<br />

to () 450 000/<br />

µl !<br />

giant forms<br />

In most cases<br />

dry tap<br />

Advanced<br />

fibrosis !<br />

Yes! No No No<br />

In the acceleration<br />

phase<br />

and blast crisis<br />

del (20q), +8,<br />

+9, +1q, and<br />

others<br />

-7, +8, +9,<br />

+1q, and others<br />

Megakaryocytes<br />

clearly<br />

elevated<br />

and<br />

arranged<br />

in nests<br />

Megakaryocytes<br />

clearly<br />

elevated,<br />

arranged<br />

in nests<br />

Very rare<br />

! Normal to Normal to<br />

<br />

Normal Normal<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


116<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Characteristics <strong>of</strong> CML<br />

Age <strong>of</strong> onset: Any age. Peak inicidence about 50 years.<br />

Clinical findings: Slowly developing fatigue, anemia; in some cases<br />

palpable splenomegaly; no fever.<br />

CBC: Leukocytosis and a left shift in the granulocyte series; possibly<br />

Hb , thrombocytes or .<br />

Advanced diagnostics: Bone marrow, cytogenetics, and molecular<br />

genetics (Philadelphia chromosome and BCR-ABL rearrangement).<br />

Differential diagnosis: Reactive leukocytoses (alkaline phosphatase,<br />

trigger?); other myeloproliferative disorders (bone marrow, cytogenetics,<br />

alkaline phosphatase).<br />

Course, therapy: Chronic progression. Acute transformation after<br />

years. New, curative drugs are currently under development. Evaluate<br />

the possibility <strong>of</strong> a bone marrow transplant (up to age approx. 60<br />

years).<br />

Steps in the Diagnosis <strong>of</strong> Chronic Myeloid Leukemia<br />

Left-shift leukocytosis in conjunction with usually low-grade anemia,<br />

thrombocytopenia or thrombocytosis (which <strong>of</strong>ten correlates with the<br />

migration <strong>of</strong> small megakaryocyte nuclei into the blood stream), and clinical<br />

splenomegaly is typical <strong>of</strong> CML. LDH and uric acid concentrations are<br />

elevated as a result <strong>of</strong> the increased cell turnover.<br />

The average “typical” cell composition is as follows (in a series analyzed<br />

by Spiers): about 2% myeloblasts, 3% promyelocytes, 24% myelocytes, 8%<br />

metamyelocytes, 57% band and segmented neutrophilic granulocytes, 3%<br />

basophils, 2% eosinophils, 3% lymphocytes, and 1% monocytes.<br />

In almost all cases <strong>of</strong> CML the hematopoietic cells display a marker<br />

chromosome, an anomalously configured chromosome 22 (Philadelphia<br />

chromosome). The translocation responsible for the Philadelphia chromosome<br />

corresponds to a special fusion gene (BCR-ABL) that can be determined<br />

by polymerase chain reaction (PCR) and fluorescence in situ hybridization<br />

(FISH).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

Left shift as far as myeloblasts, proliferation <strong>of</strong> eosinophils and<br />

basophils suggest chronic myeloid leukemia (CML)<br />

b c<br />

Fig. 39 CML. a Blood analysis in chronic myeloid leukemia (chronic phase): segmented<br />

neutrophilic granulocytes (1), band granulocyte (2) (looks like a metamyelocyte<br />

after turning and folding <strong>of</strong> the nucleus), myelocyte with defective granulation<br />

(3), and promyelocyte (4). b and c Also chronic phase: myeloblast (1),<br />

promyelocyte (2), myelocyte with defective granulation (3), immature eosinophil<br />

(4), and basophil (5) (the granules are larger and darker, the nuclear chromatin<br />

denser than in a promyelocyte).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

117


118<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Bone Marrow Analysis in CML. In many clinical situations, the findings<br />

from the CBC, the BCR-ABL transformation and the enlarged spleen unequivocally<br />

point to a diagnosis <strong>of</strong> CML. Analysis <strong>of</strong> the bone marrow<br />

should be performed because it provides a series <strong>of</strong> insights into the disease<br />

processes.<br />

Normally, the cell density is considerably elevated and granulopoietic<br />

cells predominate in the CBC. Cells in this series mature properly, apart<br />

from a slight left shift in the chronic phase <strong>of</strong> CML. CML differs from reactive<br />

leukocytoses because there are no signs <strong>of</strong> stress, such as toxic granulation<br />

or dissociation in the nuclear maturation process.<br />

Mature neutrophils may occasionally show pseudo-Pelger forms (p. 43)<br />

and the eosinophilic and, especially, basophilic granulocyte counts are<br />

<strong>of</strong>ten elevated. The proportion <strong>of</strong> cells from the red blood cell series<br />

decreases. Histiocytes may store glucocerebrosides, as in Gaucher syndrome<br />

(pseudo-Gaucher cells), or lipids in the form <strong>of</strong> sea-blue precipitates<br />

(sea-blue histiocytes after Romanowsky staining).<br />

Megakaryocytes are usually increased and are <strong>of</strong>ten present as micromegakaryocytes,<br />

with one or two nuclei which are only slightly larger than<br />

those <strong>of</strong> promyelocytes. Their cytoplasm typically shows clouds <strong>of</strong><br />

granules, as in the maturation <strong>of</strong> thrombocytes.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

Bone marrow analysis is not obligatory in chronic myeloid leukemia,<br />

but helps to distinguish between the various chronic myeloproliferative<br />

disorders<br />

b c<br />

Fig. 40 Bone marrow cytology in CML. a Bone marrow cytology in the chronic<br />

phase: increased cell density due to increased, left-shifted granulopoiesis, e.g.,<br />

promyelocyte nest (1) and megakaryopoiesis (2). Eosinophils are increased (arrows),<br />

erythropoiesis reduced. b Often micromegakaryocytes are found in the<br />

bone marrow cytology. c Pseudo-Gaucher cells in the bone marrow in CML.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

119


120<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Blast Crisis in Chronic Myeloid Leukemia<br />

During the course <strong>of</strong> CML with or without therapy, regular monitoring <strong>of</strong><br />

the differential smear is particularly important, since over periods <strong>of</strong> varying<br />

duration the relative proportions <strong>of</strong> blasts and promyelocytes increases<br />

noticeably. When the blast and promyelocyte fractions together<br />

make up 30%, and at the same time Hb has decreased to less than 10 g/dl<br />

and the thrombocyte count is less than 100 000/µl, an incipient acute blast<br />

crisis must be assumed. this blast crisis is <strong>of</strong>ten accompanied or preceded<br />

by a markedly increased basophil count. Further blast expansion—usually<br />

largely recalcitrant to treatment—leads to a clinical picture not always<br />

clearly distinguishable from acute leukemia. If in the chronic phase the<br />

disease was “latent” and medical treatment was not sought, enlargement<br />

<strong>of</strong> the spleen, slight eosinophilia and basophilia, and the occasional presence<br />

<strong>of</strong> normoblasts, together with the overwhelming myeloblast fraction,<br />

are all signs indicating CML as the cause <strong>of</strong> the blast crisis.<br />

As in AML, in two-thirds <strong>of</strong> cases cytological and immunological tests<br />

are able to identify the blasts as myeloid. In the remaining one-third <strong>of</strong><br />

cases, the cells carry the same markers as cells in ALL. This is a sign <strong>of</strong> dedifferentiation.<br />

A final megakaryoblastic or a final erythremic crisis is extremely<br />

rare.<br />

Bone marrow cytology is particularly indicated when clinical symptoms<br />

such as fatigue, fever, and painful bones suggest an acceleration <strong>of</strong> CML<br />

which is not yet manifest in the CBC. In such a case, bone marrow analysis<br />

will frequently show a much more marked shift to blasts and promyelocytes<br />

than the CBC. A proportion <strong>of</strong> more than 20% immature cell fractions<br />

is sufficient to diagnose a blast crisis.<br />

The prominence <strong>of</strong> other cell series (erythropoiesis, thrombopoiesis) is<br />

reduced. The basophil count may be elevated.<br />

A bone marrow aspiration may turn out to be empty (sicca) or scarcely<br />

yield any material. This suggests fibrosis <strong>of</strong> the bone marrow, which is<br />

frequently a complicating symptom <strong>of</strong> long-standing disease. Staining <strong>of</strong><br />

the fibers will demonstrate this condition in the bone marrow histology.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

In the course <strong>of</strong> chronic myeloid leukemia, an acute crisis may<br />

develop in which blasts predominate<br />

b c<br />

Fig. 41 Acute blast crisis in CML. a Myeloblasts (1) with somewhat atypical nuclear<br />

lobes. Basophilic granulocyte (2) and band granulocyte (3). Thrombocytopenia.<br />

The proliferation <strong>of</strong> basophilic granulocytes <strong>of</strong>ten precedes the blast crisis.<br />

b Myeloblasts in an acute CML blast crisis. Typical sand-like chromatin structure<br />

with nucleoli. A lymphocyte. c Bone marrow cytology in acute CML blast crisis:<br />

blasts <strong>of</strong> variable sizes around a hyperlobulated megakaryocyte (in this case during<br />

a lymphatic blast crisis).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

121


122<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Osteomyelosclerosis<br />

When anemia accompanied by moderately elevated (although sometimes<br />

reduced) leukocyte counts, thrombocytopenia or thrombocytosis, clinically<br />

evident splenic tumor, left shift up to and including sporadic myeloblasts,<br />

and eosinophilia, the presence <strong>of</strong> a large proportion <strong>of</strong> red cell precursors<br />

(normoblasts) in the differential blood analysis, osteomyelosclerosis<br />

should be suspected. BCR-ABL gene analysis is negative.<br />

Pathologically, osteomyelosclerosis usually originates from megakaryocytic<br />

neoplasia in the bone marrow and the embryonic hematopoietic<br />

organs, particularly spleen and liver, accompanied by fibrosis<br />

(= sclerosis) that will eventually predominate in the surrounding tissue.<br />

The central role <strong>of</strong> cells <strong>of</strong> the megakaryocyte series is seen in the giant<br />

thrombocytes, or even small coarsely structured megakaryocyte nuclei<br />

without cytoplasm, that migrate into the blood stream and appear in the<br />

CBC. OMS can be a primary or secondary disease. It may arise during the<br />

course <strong>of</strong> other myeloproliferative diseases (<strong>of</strong>ten polycythemia vera or<br />

idiopathic thrombocythemia).<br />

Tough, fibrous material hampers the sampling <strong>of</strong> bone marrow material,<br />

which rarely yields individual cells. This in itself contributes to the<br />

bone marrow analysis, allowing differential diagnosis versus reactive fibroses<br />

(parainfectious, paraneoplastic).<br />

Characteristics <strong>of</strong> OMS<br />

Age <strong>of</strong> onset: Usually older than 50 years.<br />

Clinical findings: Signs <strong>of</strong> anemia, sometimes skin irritation, drastically<br />

enlarged spleen.<br />

CBC: Usually tricytopenia, normoblasts, and left shift.<br />

Further diagnostic procedures: Fibrous bone marrow (bone marrow<br />

histology), when appropriate and BCR-ABL (always negative).<br />

Differential diagnosis: Splenomegaly in cases <strong>of</strong> lymphadenoma or<br />

other myeloproliferative diseases: bone marrow analysis.<br />

Myel<strong>of</strong>ibrosis in patients with metastatic tumors or inflammation:<br />

absence <strong>of</strong> splenomegaly.<br />

Course, therapy: Chronic disease progression; transformation is rare.<br />

If there is splenic pressure: possibly chemotherapy, substitution therapy.<br />

Further myeloproliferative diseases are described together with the relevant<br />

cell systems: polycythemia vera (see p. 162) and essential thrombocythemia<br />

(see p. 170).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Enlarged spleen and presence <strong>of</strong> immature white cell precursors<br />

in peripheral blood suggest osteomyelosclerosis<br />

Fig. 42 Osteomyelosclerosis (OMS). a and b Screening <strong>of</strong> blood cells in OMS: red<br />

cell precursors (orthochromatic erythroblast = 1 and basophilic erythroblast = 2),<br />

basophilic granulocyte (3), and teardrop cells (4). c Sometimes small, dense<br />

megakaryocyte nuclei are also found in the blood stream in myeloproliferative<br />

diseases. d Blast crisis in OMS: myeloblasts and segmented basophilic granulocytes<br />

(1).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

123<br />

b<br />

d


124<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

Elevated Eosinophil and Basophil Counts<br />

In accordance with their physiological role, an increase in eosinophils<br />

( 400/µl, i.e. for a leukocyte count <strong>of</strong> 6000, more than 8% in the differential<br />

blood analysis) is usually due to parasitic attack (p. 5). In the Western<br />

hemisphere, parasitic infestations are investigated on the basis <strong>of</strong> stool<br />

samples and serology.<br />

Strongyloides stercoralis in particular causes strong, sometimes extreme,<br />

elevation <strong>of</strong> eosinophils (may be up to 50%). However, eosinophilia <strong>of</strong><br />

variable degree is also seen in ameba infection, in lambliasis (giardiasis),<br />

schistosomiasis, filariasis, and even malaria.<br />

Bacterial and viral infections are both unlikely ever to lead to eosinophilia<br />

except in a few patients with scarlet fever, mononucleosis, or infectious<br />

lymphocytosis. The second most common group <strong>of</strong> causes <strong>of</strong> eosinophilia<br />

are allergic conditions: these include asthma, hay fever, and various dermatoses<br />

(urticaria, psoriasis). This second group also includes druginduced<br />

hypersensitivity with its almost infinitely multifarious triggers,<br />

among which various antibiotics, gold preparations, hydantoin derivatives,<br />

phenothiazines, and dextrans appear to be the most prevalent.<br />

Eosinophilia is also seen in autoimmune diseases, especially in<br />

scleroderma and panarteritis. All neoplasias can lead to “paraneoplastic”<br />

eosinophilia, and in Hodgkin’s disease it appears to play a special role in<br />

the pathology, although it is nevertheless not always present.<br />

A specific hypereosinophilia syndrome with extreme values (usually<br />

40%) is seen clinically in association with various combinations <strong>of</strong><br />

splenomegaly, heart defects, and pulmonary infiltration (Loeffler syndrome),<br />

and is classified somewhere between autoimmune diseases and<br />

myeloproliferative syndromes. Of the leukemias, CML usually manifests<br />

moderate eosinophilia in addition to its other typical criteria (see p. 114).<br />

When moderate eosinophilia dominates the hematological picture, the<br />

term chronic eosinophilic leukemia is used. Acute, absolute predominance<br />

<strong>of</strong> eosinophil blasts with concomitant decrease in neutrophils, erythrocytes,<br />

and thrombocytes suggests the possibility <strong>of</strong> the very rare acute<br />

eosinophilic leukemia.<br />

Elevated Basophil Counts. Elevation <strong>of</strong> segmented basophils to more than<br />

2–3% or 150/µl is rare and, in accordance with their physiological role in<br />

the immune system regulation, is seen inconsistently in allergic reactions<br />

to food, drugs, or parasites (especially filariae and schistosomes), i.e., usually<br />

in conditions in which eosinophilia is also seen. Infectious diseases<br />

that may show basophilia are tuberculosis and chickenpox; metabolic diseases<br />

where basophilia may occur are myxedema and hyperlipidemia. Autonomic<br />

proliferations <strong>of</strong> basophils are part <strong>of</strong> the myeloproliferative<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

b<br />

Eosinophilia and basophilia are usually accompanying phenomena<br />

in reactive and myeloproliferative disorders, especially<br />

CML<br />

c d<br />

Fig. 43 Eosinophilia and basophilia. a Screening view <strong>of</strong> blood cells in reactive<br />

eosinophilia: eosinophilic granulocytes (1), segmented neutrophilic granulocyte<br />

(2), and monocyte (3) (reaction to bronchial carcinoma). b and c The image shows<br />

an eosinophilic granulocyte (1) and a basophilic granulocyte (2) (clinical osteomyelosclerosis).<br />

d Bone marrow in systemic mastocytosis: tissue mast cell (3),<br />

which, in contrast to a basophilic granulocyte, has an unlobed nucleus, and the cytoplasm<br />

is wide with a tail-like extension. Tissue mast cells contain intensely basophilic<br />

granules.<br />

125<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


126<br />

Table 21 Different forms <strong>of</strong> benign and malignant proliferation <strong>of</strong> tissue mast<br />

cells<br />

Clinical picture Clinical diagnosis Evidence<br />

In childhood, solitary or<br />

multiple skin nodules,<br />

some brownish<br />

Sometimes transition<br />

Diffuse brownish papules,<br />

with urticaria on irritation<br />

<br />

Hyperpigmented spots,<br />

papules and/or dermographism;<br />

histamine<br />

symptoms: flushing, headache,<br />

pruritus, abdominal<br />

spasms, shock<br />

<br />

Malignant transformation,<br />

possibly with osteolysis,<br />

enlarged lymph nodes,<br />

splenomegaly, hepatomegaly<br />

<br />

Abnormalities <strong>of</strong> the White Cell Series<br />

pathologies and can develop to the extent <strong>of</strong> being termed “chronic basophilic<br />

leukemia.” In the very rare acute basophilic leukemia, the cells in<br />

the basophilic granulocyte lineage mature in the bone marrow only to the<br />

stage <strong>of</strong> promyelocytes, giving a picture similar to type M3 AML (p. 98).<br />

Being an expression <strong>of</strong> idiopathic disturbance <strong>of</strong> bone marrow function,<br />

elevated basophil counts are a relatively constant phenomenon in myeloproliferative<br />

syndromes (in addition to the specific signs <strong>of</strong> these diseases),<br />

especially in CML. Acute basophilic leukemia is extremely rare; in this<br />

condition, some <strong>of</strong> the dedifferentiated blasts contain more or less basophilic<br />

granules.<br />

The tissue-bound analogs <strong>of</strong> the segmented basophils, the tissue mast<br />

cells, can show benign or malignant cell proliferation, including the (extremely<br />

rare) acute mast cell leukemia (Table 21).<br />

Migration <strong>of</strong> leukemic cells<br />

to peripheral blood<br />

Localized mastocytosis Histology<br />

Urticaria pigmentosa Typical clinical<br />

picture<br />

Systemic mastocytosis Histology<br />

Malignant mastocytosis*<br />

Acute mast cell<br />

leukemia<br />

Histology<br />

* May occur de novo without preceding stages and without skin involvement.<br />

CBC (bone marrow)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Erythrocyte and Thrombocyte<br />

Abnormalities<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


128<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Clinically Relevant Classification Principle for Anemias:<br />

Mean Erythrocyte Hemoglobin Content (MCH)<br />

In current diagnostic practice, erythrocyte count and hemoglobin content<br />

(grams per 100 ml) in whole blood are determined synchronously. This allows<br />

calculation <strong>of</strong> the hemoglobin content per individual erythrocyte<br />

(mean corpuscular hemoglobin, MCH) using the following simple formula<br />

(p. 10):<br />

Hb (g/dl) · 10<br />

Ery (10 6 /µl)<br />

The mean cell volume, hematocrit, MCH, and erythrocyte size can be used<br />

for various calculations (Table 22; methods p. 10, normal values Table 2,<br />

p. 12). Despite this multiplicity <strong>of</strong> possible measures, however, in routine<br />

diagnostic practice the differential diagnosis in cases <strong>of</strong> low Hb concentration<br />

or low erythrocyte counts relies above all on the MCH, and most<br />

forms <strong>of</strong> anemia can safely be classified by reference to the normal data<br />

range <strong>of</strong> 26–32 pg Hb/cell (1.61–1.99 fmol/cell) as normochromic (within<br />

the normal range), hypochromic (below the), or hyperchromic (above the<br />

norm). The reticulocyte count (p. 11) provides important additional<br />

pathophysiological information. Anemias with increased erythrocyte production<br />

(hyper-regenerative anemias) suggest a high reticulocyte count,<br />

while anemias with diminished erythrocyte production (hyporegenerative<br />

anemias) have low reticulocyte counts (Table 22).<br />

It should be noted that hyporegenerative anemias due to iron or vitamin<br />

deficiency can rapidly display hyper-regeneration activity after<br />

only a short course <strong>of</strong> treatment with iron or vitamin supplements (up to<br />

the desirable “reticulocyte crisis”).<br />

The practical classification <strong>of</strong> anemia starts with the MCH:<br />

— 26–32 pg = normochromic<br />

— Less than 26 pg = hypochromic<br />

— More than 32 pg = hyperchromic<br />

Hypochromic Anemias<br />

Iron Deficiency Anemia<br />

Most anemias are hypochromic. Their usual cause is iron deficiency from<br />

various causes (Fig. 44). To distinguish quickly between real iron deficiency<br />

and an iron distribution disorder, iron and ferritin levels should be<br />

determined.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Insufficient iron absorption:<br />

● Lower than normal acidity, no<br />

acidity, stomach resection,<br />

accelerated passage from<br />

stomach to intestines<br />

● Substances that inhibit absorption<br />

<strong>of</strong> iron, such as citric<br />

acids and lactic acids, mucus and<br />

similar materials<br />

● Substances that facilitate iron<br />

absorption are missing<br />

(e.g. vitamin C)<br />

Insufficient iron<br />

supplementation:<br />

● Iron-deficient diet<br />

● In newborns: insufficient<br />

iron transfer from the<br />

mother during gravidity<br />

Endogenous redistribution:<br />

● Tumors, infections, lung<br />

hemosiderosis<br />

Iron deficiency:<br />

Hypochromic Anemias<br />

129<br />

Chronic bleeding and pathological<br />

loss <strong>of</strong> iron in the following diseases:<br />

● Disorders <strong>of</strong> the stomach and<br />

intestines (positive benzidine<br />

test)<br />

● Diseases <strong>of</strong> the urethra<br />

(hematuria!)<br />

● Diseases <strong>of</strong> the female reproductive<br />

cycle (menorrhagia,<br />

metrorrhagia)<br />

Increased iron requirement:<br />

● Growth/maturation<br />

● Increased production <strong>of</strong> the<br />

new blood cells<br />

Physiological iron loss<br />

in females:<br />

● Menses<br />

● Lactation<br />

● Pregnancy<br />

● Fatigue, koilonychia, infectious angle<br />

(angulus infectiosus oris), Plummer-<br />

Vinson syndrome, possibly “iron<br />

deficiency fever”<br />

● Anemia<br />

● Hypochromy, ring-shaped erythrocytes<br />

● Light serum, deficient in color<br />

components<br />

● Serum iron very reduced<br />

Fig. 44 The most important reasons for iron deficiency (according to<br />

Begemann)<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


130<br />

Table 22 Diagnostic findings and work-up for the most important disorders <strong>of</strong><br />

the red cell series<br />

Clinical<br />

findings<br />

Fatigue,<br />

pallor<br />

(dysphagia)<br />

Possibly<br />

fever, weight<br />

loss<br />

Diffuse<br />

symptoms<br />

Hb MCH Erythrocytemorphology<br />

Ring-shaped<br />

erythrocytes,<br />

anisocytosis<br />

Anisocytosis,<br />

poikilocytosis<br />

/n/ All sizes,<br />

dimorphic<br />

Reticulocytes<br />

Leukocytes<br />

Segmented<br />

nuclei<br />

(%)<br />

Lymphocytes<br />

(%)<br />

Other cells Thrombocytes<br />

n n n - n/<br />

n/ n n Possibly<br />

eosinophils<br />

<br />

monocytes <br />

left shift<br />

n<br />

n/ n n - n/<br />

Acute<br />

hemorrhage<br />

n n n n () () - n//<br />

Subjaundice n n or pathol., n/ n n Possibly nor-<br />

possibly<br />

spherocytes<br />

moblasts<br />

Spleen Target cells n n n Possibly normoblasts<br />

Paller n n Possibly <br />

possibly<br />

infections<br />

and bleeding<br />

tendency<br />

monocytes<br />

Pallor<br />

possibly<br />

alcohol history<br />

Plethora,<br />

possibly<br />

spleen<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Macrocytes,<br />

megalocytes<br />

n/<br />

Possibly<br />

hypersegmented<br />

granulocytes,<br />

possibly normoblasts<br />

/n<br />

n n n n/ n n - n/<br />

Acute bleed- <br />

ing tendency<br />

n n () n n n - <br />

Diagnostic steps proceed from left to right. ❘ The next step is usually unnecessary; : . the next<br />

step is optional; the next step is obligatory. n = normal value, = lower than normal, = ele-<br />

vated, ( ) = test not relevant, BSG = erythrocyte sedimentation rate.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

n


BSG Electrophoresis<br />

Iron Ferritin<br />

and<br />

others<br />

n n Ferritin<br />

<br />

? α2<br />

γ<br />

Ferritin<br />

n/<br />

Transferrin<br />

Tentative<br />

diagnosis<br />

Iron deficiency<br />

anemia<br />

Evidence/<br />

further diagnostics<br />

Determine<br />

source <strong>of</strong> bleeding;<br />

check iron<br />

absorption<br />

Acquired/sec- Search for<br />

ondary anemia trigger<br />

(infectious/<br />

toxic, paraneoplastic)<br />

n n n/ Sideroachrestic<br />

anemia,<br />

myelodysplasia<br />

n n n/ – n/ Anemia due to<br />

hemorrhaging<br />

n n n Haptoglobulin<br />

<br />

n n n Haptoglobulin<br />

<br />

(n/) Hemolytic<br />

anemia<br />

Especially:<br />

thalassemia<br />

n /(n) n/ (/n) Aplastic anemia<br />

or bone<br />

marrow carcinosia<br />

n n n/ Vitamin<br />

B12<br />

folic acid<br />

<br />

() Megaloblastic<br />

anemia<br />

Search for<br />

source and<br />

reason for<br />

hemorrhages<br />

Osmotic<br />

resistance,<br />

Coombs test,<br />

Hb electrophoresis<br />

and<br />

further tests<br />

Search for trigger<br />

or tumor<br />

Gastroscopy,<br />

determination<br />

<strong>of</strong> antibodies,<br />

possibly<br />

Schilling test<br />

n/ n Polyeythemia, ALP, progress-<br />

DD: hypergamionmaglobulinemia n n duration<br />

<strong>of</strong><br />

hemorrhaging<br />

<br />

PTT n<br />

– Thrombocytopenic<br />

purpura<br />

(ITP)<br />

Hypochromic Anemias<br />

Search for triggers,<br />

possibly<br />

also for antibodies<br />

.<br />

Bone marrow Ref.<br />

page<br />

Erythropoiesis <br />

sideroblasts ,<br />

iron in macrophages<br />

<br />

Erythropoiesis ,<br />

sideroblasts <br />

iron in macrophages<br />

131<br />

p. 132<br />

p. 134<br />

.<br />

Erythropoiesis , p. 106<br />

ring sideroblasts<br />

present<br />

.<br />

(Erythropoiesis<br />

)<br />

p. 140<br />

(Erythropoiesis p. 140<br />

, right shift)<br />

increased storage<br />

<strong>of</strong> iron<br />

p. 138<br />

Hypoplasia <strong>of</strong> all<br />

cell series, or carcinoma<br />

cells<br />

Erythropoietic<br />

megaloblasts<br />

p. 146,<br />

148, 150<br />

p. 152<br />

.<br />

Erythropoiesis p. 162<br />

Elevated megakaryocyte<br />

count<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

p. 166


132<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Table 23 Normal ranges for physiological iron and its transport proteins<br />

Old units SI units<br />

Serum iron<br />

Newborns 150–200µg/dl 27–36µmol/l<br />

Adults<br />

Female 60–140µg/dl 11–25µmol/l<br />

Male 80–150µg/dl 14–27µmol/l<br />

TIBC 300–350µg/dl 54–63µmol/l<br />

Transferrin 250–450µg/dl 2.5–4.5 g/l<br />

Serum ferritin 30–300µg/l<br />

(15–160µg/l premenopausal)<br />

TIBC = Total iron binding capacity.<br />

This usually renders determination <strong>of</strong> transferrin and total iron binding<br />

capacity (TIBC) unnecessary. If samples are being sent away to a laboratory,<br />

it is preferable to send serum produced by low-speed centrifugation,<br />

since the erythrocytes in whole blood can become mechanically damaged<br />

during shipment and may then release iron. Table 23 shows the variation<br />

<strong>of</strong> serum iron values according to gender and age.<br />

It is important to note that acute blood loss causes normochromic anemia.<br />

Only chronic bleeding or earlier serious acute blood loss leads to iron<br />

deficiency manifested as hypochromic anemia.<br />

Iron Deficiency and Blood Cell Analysis Focusing on the erythrocyte morphology<br />

is the quickest and most efficient way to investigate hypochromic<br />

anemia when the serum iron has dropped below normal values. In hypochromic<br />

anemia with iron and hemoglobin deficiency (whether due to insufficient<br />

iron intake or an increased physiological iron requirement),<br />

erythrocyte size and shape does not usually vary much (see Fig. 45). Only in<br />

advanced anemias (from approx. 11 g/dl, equivalent to 6.27 mmol/l Hb)<br />

are relatively small erythrocytes (microcytes) with reduced MCV and<br />

MCH and grayish stained basophilic erythrocytes (polychromatic erythrocytes)<br />

seen, indicating inadequate hemoglobin content. Cells with the<br />

appearance <strong>of</strong> relatively large polychromatic erythrocytes are reticulocytes.<br />

The details <strong>of</strong> their morphology can be seen after supravital staining<br />

(p. 141). A few target cells (p. 139) will be seen in conditions <strong>of</strong> severe iron<br />

deficiency.<br />

In severe hemoglobin deficiency ( 8 g/dl, equivalent to 4.96 mmol/l)<br />

the residual hemoglobin is found mostly at the peripheral edge <strong>of</strong> the<br />

erythrocyte, giving the appearance <strong>of</strong> a ring-shaped erythrocyte.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Small, hemoglobin-poor erythrocytes indicate iron deficiency<br />

Fig. 45 Iron deficiency anemia. a and b Erythrocyte morphology in iron deficiency<br />

anemia: ring-shaped erythrocytes (1), microcytes (2) faintly visible target cells<br />

(3), and a lymphocyte (4) for size comparison. Normal-sized erythrocytes (5) after<br />

transfusion. c Bone marrow cytology in iron deficiency anemia shows only increased<br />

hematopoiesis and left shift to basophilic erythroblasts (1). d Absence <strong>of</strong><br />

iron deposits after iron staining (Prussian blue reaction). Megakaryocyte (1).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

133<br />

b<br />

d


134 Erythrocyte and Thrombocyte Abnormalities<br />

Hypochromic Infectious or Toxic Anemia<br />

(Secondary Anemia)<br />

Among the various causes <strong>of</strong> lack <strong>of</strong> iron for erythropoiesis (see Fig. 44,<br />

p. 129), a special situation is represented by the internal iron shift caused<br />

by “iron pull” <strong>of</strong> the reticuloendothelial system (RES) during infections,<br />

toxic processes, autoimmune diseases, and tumors. Since this anemia results<br />

from another disorder, it is also called secondary anemia. The MCH is<br />

hypochromic, or in rare instances, normochromic, and therefore erythrocyte<br />

morphology is particularly important to diagnosis. In contrast to exogenous<br />

iron deficiency anemias, the following phenomena are <strong>of</strong>ten observed,<br />

depending on the severity <strong>of</strong> the underlying condition:<br />

➤ Anisocytosis, i.e., strong variations in the size <strong>of</strong> the erythrocytes, beyond<br />

the normal distribution. The result is that in almost every field<br />

view, some erythrocytes are either half the size or twice the size <strong>of</strong> their<br />

neighbors.<br />

➤ Poikilocytosis, i.e., variations in the shape <strong>of</strong> the erythrocytes. In addition<br />

to the normal round shape, numbers <strong>of</strong> oval, or pear, or tear shaped<br />

cells are seen.<br />

➤ Polychromophilia, the third phenomenon in this series <strong>of</strong> nonspecific<br />

indicators <strong>of</strong> disturbed erythrocyte maturation, refers to light grayblue<br />

staining <strong>of</strong> the erythrocytes, indicating severely diminished<br />

hemoglobin content <strong>of</strong> these immature cells.<br />

➤ Basophilic cytoplasmic stippling in erythrocytes is a sign <strong>of</strong> irregular regeneration<br />

and <strong>of</strong>ten occurs nonspecifically in secondary anemia.<br />

The reticulocyte count is usually reduced in infectious or toxic anemia, unless<br />

there is concomitant hemolysis or acute blood loss.<br />

Bone marrow analysis in secondary anemia usually shows reduced<br />

erythropoiesis and granulopoiesis with a spectrum <strong>of</strong> immature cells (“infectious/toxic<br />

bone marrow”). The information is so nonspecific that usually<br />

bone marrow aspiration is not performed. So long as all other laboratory<br />

methods are employed, bone marrow cytology is very rarely needed<br />

in cases <strong>of</strong> hypochromic anemia.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Hypochromic erythrocytes <strong>of</strong> very variable morphology indicate<br />

secondary anemia, usually in cases <strong>of</strong> infectious disease or<br />

tumor<br />

a b<br />

Fig. 46 Secondary anemia. a and b Erythrocyte morphology in secondary hypochromic<br />

anemia: the erythrocytes vary greatly in size (anisocytosis) and shape (1)<br />

(poikilocytosis), and show basophilic stippling (2). Burr cell (3), which has no specific<br />

diagnostic significance. Occasionally, the erythrocytes stain a s<strong>of</strong>t gray–blue<br />

(4) (polychromasia). c Bone marrow cell overview in secondary anemia. Cell<br />

counts in the white cell series are elevated (promyelocytes = 1), eosinophils (2),<br />

and plasma cells (3); erythropoiesis is reduced (4).<br />

135<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

c


136 Erythrocyte and Thrombocyte Abnormalities<br />

Bone Marrow Cytology in the Diagnosis <strong>of</strong> Hypochromic<br />

Anemias<br />

So long as all other laboratory methods are employed, bone marrow cytology<br />

is very rarely needed in cases <strong>of</strong> hypochromic anemia.<br />

Bone marrow cytology is rarely strictly indicated after all other available<br />

diagnostic methods have been exhausted (Table 22, p. 130). However, in<br />

doubtful cases it can usually at least help to rule out malignant disease.<br />

In iron deficiency anemias <strong>of</strong> the most various etiologies, erythropoiesis<br />

is stimulated in a compensatory fashion, and the distribution <strong>of</strong> the<br />

markers shows the expected increase in red cell precursors. The erythropoiesis<br />

to granulopoiesis ratio increases in favor <strong>of</strong> erythropoiesis from<br />

1 :3 to 1 :2, but rarely further. The red cell series shows a left shift, i.e.,<br />

there are more immature red cell precursors (erythroblasts and proerythroblasts)<br />

(for normal values, see Table 4). Usually, these red cell precursors<br />

do not show any clearly atypical morphology, but the cytoplasm is basophilic<br />

even in normoblasts, according with the poor hemoglobinization.<br />

Iron staining <strong>of</strong> the bone marrow shows no sideroblasts (normoblasts containing<br />

iron granules), or only very few ( 10%, norm 30–40%). A constant<br />

finding in anemia stemming from exogenous iron deficiency is absence <strong>of</strong><br />

iron in the macrophages <strong>of</strong> the bone marrow reticulum.<br />

Megakaryocyte counts are almost always increased in iron deficiency<br />

due to chronic hemorrhaging, but can also show increased proliferation in<br />

iron deficiency from other causes (which can lead to increased thrombocyte<br />

counts in states <strong>of</strong> iron deficiency).<br />

In infectious or toxic (secondary) anemia, unlike exogenous iron deficiency<br />

anemia, erythropoiesis tends to be somewhat suppressed. There is<br />

no left shift and no specific anomalies are present. Granulopoiesis predominates<br />

and <strong>of</strong>ten shows nonspecific “stress phenomena” and a dissociation<br />

<strong>of</strong> nuclear and cytoplasmic maturation (e.g., cytoplasm that is still<br />

basophilic with promyelocytic granules in mature, banded myelocyte nuclei).<br />

Depending on the trigger <strong>of</strong> the anemia, the monocyte, lymphocyte, or<br />

plasma cell counts are <strong>of</strong>ten moderately increased, and megakaryocyte<br />

counts are occasionally slightly elevated. The important indicator is iron<br />

staining <strong>of</strong> the bone marrow. The “iron pull” <strong>of</strong> the RES leads to intensive<br />

iron storage in macrophages, while the red cell precursors are almost ironfree.<br />

However, combinations do exist, when a pre-existing iron deficiency<br />

means that the iron depositories are empty even in an infectious or toxic<br />

process. Moreover, not every secondary anemia is hypochromic. Where<br />

there is concomitant alcoholism or vitamin deficiency, secondary anemia<br />

may be normochromic or hyperchromic.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Hypochromic Anemias<br />

Hypochromic Sideroachrestic Anemias<br />

(Sometimes Normochromic or Hyperchromic)<br />

137<br />

In a sideroachrestic anemia existing iron cannot be utilized (achrestic<br />

= useless). This is a suspected diagnosis when serum iron levels are<br />

raised or in the high normal range, and when the erythrocytes show strong<br />

anisocytosis, poikilocytosis (with reduced average MCV), polychromophilia,<br />

and in some cases also basophilic stippling (Fig. 46). This suspicion can be<br />

further illuminated by bone marrow analysis. Unlike in infectious/toxic<br />

anemias, the red cell series is well represented. Iron staining <strong>of</strong> the bone<br />

marrow is the decisive diagnostic test, causing the iron-containing red<br />

precursor cells (sideroblasts) to stand out (hence the term “sideroblastic<br />

anemia.”) The iron precipitates <strong>of</strong>ten collect in a ring around the nucleus<br />

(“ringed sideroblasts”).<br />

By far the majority <strong>of</strong> the “idiopathic sideroachrestic anemias,” as they<br />

used to be called, are myelodysplasias (see p. 106). Only a few <strong>of</strong> them appear<br />

to be hereditary or have exogenous triggers (alcoholism, lead poisoning).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


138 Erythrocyte and Thrombocyte Abnormalities<br />

Hypochromic Anemia with Hemolysis<br />

Thalassemias<br />

A special form <strong>of</strong> hypochromic anemia mostly affecting patients <strong>of</strong> Mediterranean<br />

descent presents with normal erythrocyte count, decreased<br />

MCH, and clinical splenomegaly. The smear displays erythrocytes with<br />

central hemoglobin islands (target cells). These cells do not necessarily<br />

predominate in the CBC: the most revealing field views show at most 50%<br />

target cells in addition to clear anisocytosis and frequent basophilic stippling.<br />

Occasional normoblasts give a general indication <strong>of</strong> increased erythropoiesis.<br />

Although target cells are also nonspecific, since they can occur<br />

in such conditions as severe iron deficiency or obstructive jaundice, this<br />

overall picture should prompt hemoglobin electrophoresis. The sample<br />

consists <strong>of</strong> ACD-stabilized blood at 1 :10 dilution. A significant increase in<br />

the HbA2 fraction confirms a diagnosis <strong>of</strong> thalassemia minor, the heterozygous<br />

form <strong>of</strong> the disease. Thalassemia major, the homozygous variant, is<br />

far rarer and more serious. In this form <strong>of</strong> the disease, in addition to the<br />

target cells, the CBC shows a marked increase in red precursor cells. Hbelectrophoresis<br />

shows a predominance <strong>of</strong> HbF (the other hemolytic anemias<br />

are usually normochromic, see p. 140).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Hypochromic anemia without iron deficiency, sometimes with<br />

target cells, suggests thalassemia<br />

a b<br />

Fig. 47 Thalassemia. a Thalassemia minor: <strong>of</strong>ten no target cells, but an increase<br />

in the number <strong>of</strong> small erythrocytes (shown here in comparison with a lymphocyte),<br />

so that sometimes there is no anemia. b More advanced thalassemia minor:<br />

strong anisocytosis and poikilocytosis (1), basophilic stippling (2), and sporadic<br />

target cells (3). c Thalassemia major: erythroblasts (1), target cell (2), polychromatic<br />

erythrocytes (3), and Howell–Jolly bodies (4) (in a case <strong>of</strong> functional asplenia).<br />

Lymphocyte (5) and granulocyte (6).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

139<br />

c


140 Erythrocyte and Thrombocyte Abnormalities<br />

Normochromic Anemias<br />

Anemias where red cell hemoglobinization is normal (26–32 pg/dl, equivalent<br />

to 1.61–1.99 fmol/l) and average MCVs are normal (77–100 fl) can<br />

broadly be explained by three mechanisms: a) acute blood loss with sufficient<br />

metabolic reserves remaining; b) elevated cell turnover in which iron<br />

is reused as soon as it becomes free, so that hypochromia does not arise<br />

(this is typical <strong>of</strong> almost all hemolytic anemias except thalassemias; see<br />

p. 138); and c) suppression <strong>of</strong> cell production under conditions <strong>of</strong> normal<br />

iron supply (this is the group <strong>of</strong> hypoplastic–aplastic anemias, which have<br />

a variety <strong>of</strong> causes).<br />

— In cases <strong>of</strong> acute blood loss: clinical findings, occult blood?<br />

— In hemolytic cases: reticulocytes , haptoglobin , possibly bilirubin<br />

.<br />

— In bone marrow suppression: e.g. aplastic anemia, reticulocytes .<br />

Normochromic Hemolytic Anemias<br />

Hemolytic anemias result from a shortened erythrocyte life span with insufficient<br />

compensation from increased erythrocyte production (Table<br />

24).<br />

Usually, hematopoiesis in the bone marrow is increased in compensation,<br />

and, depending on the course <strong>of</strong> the disease, may make up for the accelerated<br />

cell degradation for all or some <strong>of</strong> the time by recycling the iron as<br />

it becomes free.<br />

Accordingly, counts <strong>of</strong> the young, newly emerged erythrocytes (reticulocytes)<br />

are always raised, and usually sporadic normoblasts are found. Anemia<br />

proper <strong>of</strong>ten becomes apparent only in a “crisis” with acute, accelerated<br />

cell degradation, and reticulocyte counts increased up to more than<br />

500%.<br />

A common cellular phenomenon after extended duration <strong>of</strong> hemolytic<br />

anemia is the manifestation <strong>of</strong> macrocytic hypochromic disorders (p. 150),<br />

because the chronic elevation <strong>of</strong> hematopoietic activity can exhaust the<br />

endogenous folic acid reserves (pernicious anemia).<br />

Bone marrow analysis shows both relative and absolute increases in<br />

erythropoietic activity: among the red cell precursors, in acute severe<br />

hemolysis the more immature forms <strong>of</strong>ten predominate more than in normal<br />

bone marrow, and in chronic hemolysis the maturer forms do<br />

(orthochromatic normoblasts). In addition, the normoblasts in hemolytic<br />

bone marrow <strong>of</strong>ten are markedly clustered (Fig. 48), whereas in normal<br />

bone marrow they are more evenly dispersed (Fig. 18).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Consistently elevated “young” erythrocytes (reticulocytes) suggest<br />

hemolysis<br />

a b<br />

Fig. 48 Hemolytic anemia. a and b Newly formed erythrocytes appear as large,<br />

polychromatic erythrocytes (1) after Pappenheim staining (a); supravital staining<br />

(b) reveals spot-like precipitates (reticulocyte = 2). Thrombocyte (3). c Bone marrow<br />

cells in hemolytic anemia at low magnification: increased hematopoiesis with<br />

cell clusters. Orthochromatic erythroblasts predominate. A basophilic erythroblast<br />

shows loosened nuclear structure (arrow), a sign <strong>of</strong> secondary folic acid deficiency.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

141<br />

c


142<br />

Table 24 Causes <strong>of</strong> the most common hemolytic anemias<br />

Special morphological<br />

features <strong>of</strong> erythrocytes<br />

Causes within the erythrocytes (corpuscular hemolyses)<br />

➤ Hereditary<br />

Membrane abnormalities<br />

– Spherocytosis (see p. 144)<br />

– Elliptocytosis<br />

Hemoglobin abnormalities<br />

– Thalassemia (see p. 138)<br />

– Sickle cell anemia (see<br />

p. 144)<br />

– Other rare hemoglobin-<br />

related disorders<br />

Enzyme defects<br />

– Glucose-6-phosphate<br />

dehydrogenase<br />

– Pyruvate kinase and<br />

many others<br />

➤ Acquired<br />

Paroxysmal nocturnal<br />

hemoglobinuria<br />

– Small spherocytes<br />

– Elliptocytes<br />

– Target cells<br />

– Sickle cells<br />

– Possibly Heinz bodies<br />

– Macrocytes<br />

Zieve syndrome – Foam cells in the bone<br />

marrow<br />

Further advanced<br />

diagnostics<br />

Osmotic resistance<br />

Hemoglobin electrophoresis<br />

Enzyme tests<br />

Sucrose hemolysis<br />

test, absence <strong>of</strong> CD 55<br />

(DAF) and CD 59<br />

MIRL (membrane<br />

inhibitor <strong>of</strong> reactive<br />

lysis)<br />

Causes outside the erythrocytes (extracorpuscular hemolyses)<br />

➤ Biosynthesis <strong>of</strong> antibodies<br />

Isoantibodies (fetal erythro-<br />

Rh serology<br />

blastosis, transfusion events)<br />

Warm autoantibodies Coombs test<br />

Cold autoantibodies<br />

Chemical-allergic antibodies<br />

(e. g., cephalosporin, methyldopa)<br />

– Autoagglutination Coombs test,<br />

Cold agglutination<br />

titer<br />

➤ Physical or chemical noxae<br />

(e. g., after burns, heart valve<br />

replacement; heavy metal<br />

exposure, animal- or plantderived<br />

poisons)<br />

– Partially Heinz bodies<br />

➤ Microangiopathic hemolysis in<br />

hemolytic-uremic syndrome,<br />

thrombotic-thrombocytopenic<br />

purpura, bone marrow carcinoses<br />

➤ Infection-related noxae (e. g.<br />

influenza, salmonella infection,<br />

malaria)<br />

Erythrocyte and Thrombocyte Abnormalities<br />

➤ Hypersplenism, e. g. lymphatic<br />

system disease, infections with<br />

splenomegaly, portal hypertension<br />

– Schizocytes, fragmentocytes<br />

(see p. 143)<br />

– For malaria pathogen<br />

(see p. 158)<br />

Thrombocytes <br />

Liver, kidney<br />

Demonstration <strong>of</strong><br />

pathogen<br />

Cause <strong>of</strong><br />

splenomegaly<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

Distribution pattern and shape <strong>of</strong> erythrocytes can be relevant in<br />

the diagnosis <strong>of</strong> hemolysis<br />

b c<br />

Fig. 49 Autoagglutination and fragmentocytes. a Clumps <strong>of</strong> erythrocytes. If this<br />

is the picture in all regions <strong>of</strong> the smear, an artifact is unlikely and serogenic (auto)agglutination<br />

should be suspected (in this case due to cryoagglutinins in mycoplasmic<br />

pneumonia). Thrombocytes are found between the agglutinated erythrocytes.<br />

b and c Conspicuous half-moon and egg-shell-shaped erythrocytes: fragmentocytosis<br />

in microangiopathic hemolytic anemia. Fragmentocytes (1), target<br />

cell (2), and echinocytes (3) (this last has no diagnostic relevance).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

143


144 Erythrocyte and Thrombocyte Abnormalities<br />

Cytomorphological Anemias with Erythrocyte<br />

Anomalies<br />

Microspherocytosis This corpuscular form <strong>of</strong> hemolysis is characterized<br />

by dominant genetic transmission, splenomegaly, and a long uneventful<br />

course with occasional hemolytic crises. Blood analysis shows erythrocytes<br />

which appear strikingly small in comparison with leukocytes. The<br />

central light area is absent or only faintly visible, since they are spherical<br />

in cross-section rather than barbell-shaped. The abnormal size distribution<br />

can be measured in two dimensions and plotted using Price–Jones<br />

charts. Close observation <strong>of</strong> the morphology in the smear (Fig. 50) is particularly<br />

important, because automated blood analyzers will determine<br />

a normal cell volume. The extremely reduced osmotic resistance <strong>of</strong> the<br />

erythrocytes (in the NaCl dilution series) is diagnostic. Coombs test is<br />

negative.<br />

Stomatocytosis is an extremely rare hereditary condition. Stomatocytes<br />

are red cells with a median streak <strong>of</strong> pallor, giving the cells a “fish mouth”<br />

appearance. A few stomatocytes may be found in hepatic disease.<br />

Hemoglobinopathies (see also thalassemia, p. 138). Target cells are<br />

frequently present (Fig. 47).<br />

In addition to target cells, smears from patients with sickle cell anemia<br />

may show a few sickle-shaped erythrocytes, but more usually these only<br />

appear under conditions <strong>of</strong> oxygen deprivation (Fig. 50). (This can be<br />

achieved by covering a fresh blood droplet with a cover glass; a droplet <strong>of</strong><br />

2% Na2S2O4 may be added). Sickle cell anemia is a dominant autosomal recessive<br />

hemoglobinopathy and is diagnosed by demonstrating the HbS<br />

band in Hb-electrophoresis. Homozygous patients with sickle cell anemia<br />

always suffer from chronic normocytic hemolysis. If provoked by oxygen<br />

deficiency or infections, severe crises may occur with clogging <strong>of</strong> the microvasculature<br />

by aggregates <strong>of</strong> malformed erythrocytes. Patients who are<br />

heterozygous for sickle cell anemia have the sickle cell trait but do not display<br />

the disease or its symptoms. These can, however, be triggered by very<br />

low oxygen tension. Sickle cell anemia is quite <strong>of</strong>ten combined with other<br />

hemoglobinopathies, such as thalassemia.<br />

Schistocytosis (Fragmentocytosis) If, in acquired hemolytic anemia, some<br />

<strong>of</strong> the erythrocytes are fragmented and have various irregular shapes<br />

(eggshell, helmet, triangle, or crescent; Fig. 49), this may be an indication<br />

<strong>of</strong> changes in the capillary system (microangiopathy), or else <strong>of</strong> disseminated<br />

intravascular coagulopathy (DIC). Microangiopathic hemolytic anemias<br />

develop in the course <strong>of</strong> thrombotic thrombocytopenic purpura (TTP,<br />

Moschcowitz disease) and its related syndromes: in children (hemolytic<br />

uremic syndrome, HUS); in pregnant women (HELLP syndrome); or in<br />

patients with bone marrow metastases from solid tumors.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Conspicuous erythrocyte morphology in anemia: microspherocytosis<br />

and sickle cell anemia<br />

a b<br />

c d<br />

Fig. 50 Microspherocytes and sickle cells. All erythrocytes are strikingly small in<br />

comparison with lymphocytes (1) and lack a lighter center: these are microspherocytes<br />

(diameter 6 µm). Polychromatic erythrocyte (2). b Erythrocytes with an<br />

elongated rather than round lighter center: these are stomatocytes, which are rarely<br />

the cause <strong>of</strong> anemia. c Native sickle cells (1) are found only in homozygous<br />

sickle cell anemia, otherwise only target cells (2) are present. d Sickle cell test under<br />

reduced oxygen tension: almost all erythrocytes appear as sickle cells in the<br />

homozygous case presented here.<br />

145<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


146 Erythrocyte and Thrombocyte Abnormalities<br />

Normochromic Renal Anemia<br />

(Sometimes Hypochromic or Hyperchromic)<br />

Normochromic anemia should also suggest the possibility <strong>of</strong> renal insufficiency,<br />

which will always lead to anemia within a few weeks. In cases <strong>of</strong><br />

chronic renal insufficiency it is always present and may reach Hb values as<br />

low as 6 g/dl. The anemia is caused by changes in the synthesis <strong>of</strong> erythropoietin,<br />

the hormone regulating erythropoiesis; measurement <strong>of</strong> serum<br />

erythropoietin is an important diagnostic tool. Erythrocyte life span is also<br />

slightly reduced.<br />

Apart from poikilocytosis, the blood cells are morphologically unremarkable.<br />

The reticulocyte counts <strong>of</strong>ten remain normal. The bone marrow<br />

does not show any significant characteristic changes, and therefore serves<br />

no diagnostic purpose in this situation.<br />

Anemias due to renal insufficiency are usually normochromic, but hypochromic<br />

or hyperchromic forms do occur. Hypochromic anemia is an indicator<br />

<strong>of</strong> the reactive process that has led to the renal insufficiency (e.g.,<br />

pyelonephritis and glomerulonephritis), resulting in secondary hypochromic<br />

anemia. In addition, dialysis patients <strong>of</strong>ten develop iron deficiency.<br />

Chronic renal insufficiency can lead to folic acid deficiency, and<br />

dialysis therapy will reinforce this, explaining why hyperchromic anemias<br />

also occur in kidney disease.<br />

Bone Marrow Aplasia<br />

Pure Red Cell Aplasia (PRCA, Erythroblastopenia)<br />

Erythroblastopenia in the sense <strong>of</strong> a purely aplastic condition in the red<br />

cell series is extremely rare. Diamond–Blackfan anemia (congenital hypoplastic<br />

anemia) is the congenital form <strong>of</strong> this disease. Acquired acute, transient<br />

infections in adults and children are usually caused by virus infections<br />

(parvovirus B19). Chronic acquired erythroblastophthisis is<br />

frequently associated with thymoma and has an autoimmune etiology.<br />

Anemia in pure erythroblastophthisis is normochromic without significant<br />

changes in the CBC for white cells and thrombocytes. Naturally, the<br />

reticulocyte count is extremely low, close to zero.<br />

In all these anemias, the bone marrow shows well-developed granulopoiesis<br />

and megakaryopoiesis, but erythropoiesis is (more or less) entirely<br />

lacking.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Unexplained decrease in cell counts for one or more lines: the<br />

bone marrow smear may show various forms <strong>of</strong> aplasia<br />

Fig. 51 Forms <strong>of</strong> bone marrow aplasia. a Bone marrow cytology in erythroblastopenia:<br />

only activated cells <strong>of</strong> the granulopoietic series are present. The megakaryopoiesis<br />

(not shown here) show no abnormalities. b Bone marrow aplasia:<br />

hematopoiesis is completely absent: only adipocytes and stroma cells are seen.<br />

c Giant erythroblast (arrow) in the bone marrow in acute parvovirus B19 infection.<br />

d Conspicuous binuclear erythroblasts in the bone marrow <strong>of</strong> a patient with congenital<br />

dyserythropoietic anemia (type II CDA).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

147<br />

b<br />

d


148<br />

Erythrocyte and Thrombocyte Abnormalities<br />

The differential diagnosis in this context relates to very rare congenital<br />

dyserythropoietic anemias (CDA). These anemias manifest mostly in<br />

childhood or youth and may be normocytic or macrocytic. The bone marrow<br />

shows increased erythropoiesis with multinucleated erythroblasts,<br />

nuclear fragmentation, and cytoplasmic bridges. There are three types<br />

(type II carries the so-called HEMPAS antigen: hereditary multinuclearity<br />

with positive acidified serum lysis test).<br />

Aplasias <strong>of</strong> All Bone Marrow Series (Panmyelopathy,<br />

Panmyelophthisis, Aplastic Anemia)<br />

A reduction in erythrocyte, granulocyte, and thrombocyte cell counts in<br />

these series, which may progress to zero, is far more common than pure<br />

erythroblastopenia and is always acquired (except in the rare pediatric<br />

Fanconi syndrome with obvious deformities).<br />

Pathologically, this life-threatening disease is a result <strong>of</strong> damage to the<br />

hematopoietic stem cells, <strong>of</strong>ten by chemical toxins or, occasionally, viral<br />

infection. An autoimmune response <strong>of</strong> the T-lymphocytes also seems to<br />

play a role.<br />

The term “panmyelopathy” is synonymous with “aplastic anemia” in<br />

the broader sense and with “panmyelophthisis.”<br />

The CBCs show the rapid progression <strong>of</strong> normochromic anemia and<br />

greatly reduced reticulocyte counts. Granulocyte counts gradually<br />

dwindle to zero, followed by the monocytes. Thrombocytes are usually<br />

also quite severely affected.<br />

The remaining blood cells in all series appear normal, although naturally,<br />

given the presence <strong>of</strong> the noxious agents or intercurrent infections,<br />

they <strong>of</strong>ten show reactive changes (e.g., toxic granulations). The bone marrow<br />

aspirate for cytological analysis is <strong>of</strong>ten notable for poor yield <strong>of</strong><br />

material, although a completely empty aspirate (dry tap) is rare.<br />

The material obtained yields unfamiliar images in a smear (Fig. 51).<br />

Frequently, strings and patches <strong>of</strong> reticular (stroma) cells from the bone<br />

marrow predominate, which normally are barely noticed in an aspirate.<br />

There are usually no signs <strong>of</strong> phagocytosis. Aside from the reticular cells,<br />

there are isolated lymphocytes, plasma cells, tissue basophils, and macrophages.<br />

Depending on the stage in the aplastic process, there may be residual<br />

hematopoietic cells. In some instances, the whole disease process is<br />

focal. For this reason, bone marrow histology must be performed<br />

whenever the cytological findings are insufficient or dubious in cases <strong>of</strong><br />

tricytopenia <strong>of</strong> unknown cause.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Table 25 Substances, suspected or proven to cause panmyelopathy<br />

Analgesics, antirheumatic<br />

drugs<br />

Hypochromic Anemias<br />

Phenylbutazone, oxyphenbutazone, other<br />

nonsteroidal antirheumatic drugs, gold<br />

preparations, penicillamine<br />

Antibiotics Chloramphenicol, sulfonamide<br />

Anticonvulsive drugs Hydantoin<br />

Thyrostatic drugs Carbimazole/methimazole<br />

Sedatives Phenothiazines<br />

Other medications Cimetidine, tolbutamide<br />

Insecticides Hexachlorcyclohexane and other chlorinated<br />

hydrocarbons<br />

Solvents Benzene<br />

Viruses z. B. Hepatitis, CMV<br />

149<br />

Differential Diagnosis versus Reduction in Cell Counts in Several Series<br />

(Bicytopenia or Tricytopenia):<br />

➤ After thorough analysis, most cytopenias with hyperplasia <strong>of</strong> the bone<br />

marrow have to be defined as myelodysplasias (p. 106), unless they are<br />

caused by accelerated cell degeneration (e.g., in hypersplenism).<br />

➤ Cytopenia with bone marrow fibrosis points to myeloproliferative-type<br />

diseases (p. 114) or results from direct toxic or inflammatory agents.<br />

➤ Cytopenia can <strong>of</strong> course also occur after bone marrow infiltration by<br />

malignant cells (carcinoma, sarcoma), which will not be contained in<br />

every bone marrow aspirate. This is why, when the diagnosis is uncertain,<br />

histological analysis should always be carried out.<br />

➤ Cytopenia can also result from B12 or folic acid deficiency (but note the<br />

possibility <strong>of</strong> hyperchromic anemia, p. 152).<br />

➤ Another cause <strong>of</strong> cytopenia is expansion <strong>of</strong> malignant hematopoietic<br />

cells in the bone marrow. This is easily overlooked if the malignant cells<br />

do not appear in the bloodstream, as in plasmacytoma, lymphadenoma,<br />

and aleukemic leukemia (leukemia without peripheral blasts).<br />

➤ Cytopenia also develops <strong>of</strong> course after high-dose radiation or<br />

chemotherapy. In such cases it is not so much the CBC or bone marrow<br />

analysis as the exposure history that will allow the condition to be distinguished<br />

from the panmyelopathies described above.<br />

➤ Cytopenia caused by panmyelopathy mechanisms (see preceding text;<br />

triggers shown in Table 25).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


150<br />

Erythrocyte and Thrombocyte Abnormalities<br />

In unexplained anemia, thrombocytopenia, or leukocytopenia, any<br />

possible triggers (Table 25) must be discontinued or avoided. Panmyelophthisis<br />

or aplastic anemia is an acute disease that can only be overcome<br />

with aggressive treatment (glucocorticoids, cyclosporin, antilymphocyte<br />

globulin).<br />

Bone Marrow Carcinosis and Other Space-Occupying<br />

Processes<br />

Anemia resulting from bone marrow infiltration by growing, spaceoccupying<br />

tumor metastases can in principle be normochromic. However,<br />

under the indirect influence <strong>of</strong> the underlying disease, it tends more <strong>of</strong>ten<br />

to be hypochromic (secondary anemia).<br />

Normoblasts in the differential blood analysis (Fig. 9 a, p. 33) particularly<br />

suggest the possibility <strong>of</strong> bone marrow carcinosis, because their presence<br />

implies destruction <strong>of</strong> the bone marrow–blood barrier. Usually, bone marrow<br />

carcinosis leads eventually to lower counts in other cell series,<br />

especially thrombocytes.<br />

Bone metastases from malignant tumors rarely affect the bone marrow<br />

and hematopoiesis, and if they do, it is usually late. The most common<br />

metastases in bone marrow derive from small-cell bronchial carcinoma<br />

and breast cancer.<br />

In the differential diagnosis, the effects <strong>of</strong> direct bone marrow infiltration<br />

must be distinguished from phenomena caused by microangiopathic<br />

hemolytic anemia (MHA) in the presence <strong>of</strong> tumor (p. 144). Carcinosis and<br />

MHA may <strong>of</strong> course coexist.<br />

Bone marrow cytology in these situations tends to reveal a generally<br />

decreased density <strong>of</strong> hematopoietic cells and signs <strong>of</strong> reactive marrow as<br />

seen in secondary anemia (p. 134). Only in a few field views—<strong>of</strong>ten at the<br />

edge <strong>of</strong> the smear—will one occasionally encounter atypical cell elements<br />

which cannot be assigned with certainty to any <strong>of</strong> the hematopoietic blast<br />

families. The critical feature is their close arrangement in clusters. These<br />

atypical cells are at least as large as myeloblasts or proerythroblasts (e.g.,<br />

in small-cell bronchial carcinoma), usually considerably larger. Tumor<br />

type cannot be diagnosed with certainty (except, e.g., melanoma). Bone<br />

marrow histology and possibly immunohistology tests must be performed<br />

if there is any doubt, or in the case <strong>of</strong> negative cytological findings<br />

or dry tap, since the clustered, focal character <strong>of</strong> metastases naturally<br />

means that they may not be obtained in every aspirate.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Thrombocytopenia with leukocytosis and erythroblasts in the<br />

peripheral blood: consider bone marrow carcinosis<br />

a b<br />

Fig. 52 Bone marrow carcinosis. a and b Bone marrow smear at low magnification<br />

showing islands <strong>of</strong> infiltration by a homogeneous cell type (a), or, alternatively,<br />

by apparently different cell types which do, however, all display identical chromatin<br />

structure and cytoplasm: bone marrow carcinosis in breast carcinoma (a) or<br />

bronchial non-small-cell carcinoma (b). c Island <strong>of</strong> dedifferentiated cells in the<br />

bone marrow which cannot be assigned to any <strong>of</strong> the hematopoietic lineages:<br />

bone marrow carcinosis (here in a case <strong>of</strong> embryonal testicular cancer).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

151<br />

c


152<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Hyperchromic Anemias<br />

In patients with clear signs <strong>of</strong> anemia, e.g., a “sickly pallor,” atrophic lingual<br />

mucosa, and sometimes also neurological signs <strong>of</strong> bathyanesthesia<br />

(loss <strong>of</strong> deep sensibility), even just a cursory examination <strong>of</strong> the blood<br />

smear may indicate the diagnosis. Marked poikilocytosis and anisocytosis<br />

are seen, and the large size <strong>of</strong> the erythrocytes is particularly conspicuous<br />

in comparison with the lymphocytes, whose diameter they exceed (megalocytes).<br />

These are the hallmarks <strong>of</strong> macrocytic, and, with respect to bone<br />

marrow cells, usually also megaloblastic anemia, with a mean cell diameter<br />

greater than 8 µm and a cell volume (MCV) usually greater than<br />

100 µm 3 . Mean cell Hb content (MCH) is more than 36 pg (1.99 fmol) and<br />

thus indicates hyperchromic anemia.<br />

Only when there is severe pre-existing concomitant iron deficiency is a<br />

combination <strong>of</strong> macrocytic cells and hypochromic MCH possible (“dimorphic<br />

anemia”).<br />

Table 26 Most common causes <strong>of</strong> hyperchromic anemias<br />

Vitamin-B12 deficiency Folic acid deficiency<br />

Nutritional deficits, e. g. Nutritional deficits<br />

– Goat milk<br />

– Chronic abuse <strong>of</strong> alcohol<br />

– Vegetarian diet<br />

– Alcoholism<br />

Impaired absorption Impaired absorption<br />

– Genuine pernicious anemia<br />

– E.g., sprue (psilosis)<br />

– Status after gastrectomy<br />

Increased requirement<br />

– Ileum resection<br />

– Crohn disease<br />

– Pregnancy<br />

– Celiac disease, sprue (psilosis)<br />

– Hemolytic anemia<br />

– Intestinal diverticulosis<br />

Interference/antagonism<br />

– Insufficiency <strong>of</strong> the exocrine pancreas – Phenylhydantoin<br />

– Fish tapeworm<br />

– Cytostatic antimetabolic drugs<br />

– Trimethoprim (antibacterial<br />

combination drug)<br />

– Oral contrazeptives<br />

– Antidepressants<br />

– Alcohol<br />

Although other rare causes exist (Table 26), almost all patients with hyperchromic<br />

anemia suffer from vitamin B12 and/or folic acid deficiency.<br />

Since a deficiency <strong>of</strong> these essential metabolic building blocks suppresses<br />

DNA synthesis not only in erythropoiesis, but in the other cell series as<br />

well, over time more or less severe pancytopenia will develop.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Conspicuous large erythrocytes suggest hyperchromic macrocytic<br />

anemia, usually megaloblastic in the bone marrow<br />

Fig. 53 Hyperchromic anemia. a Marked anisocytosis. In addition to normalsized<br />

erythrocytes (1), macrocytes (2) and large ovoid megalocytes are seen (3).<br />

Hypersegmented granulocyte (4). b In hyperchromic anemia, red cell precursors<br />

may be released into the peripheral blood: here, a polychromatic erythroblast. c<br />

and d Bone marrow in megaloblastic anemia: slight (1) or marked (2) loosening<br />

up <strong>of</strong> the nuclear structure, in some cases with binuclearity (3). Giant forms <strong>of</strong><br />

band granulocytes and metamyelocytes (4) are <strong>of</strong>ten present.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

153<br />

b<br />

d


154<br />

Erythrocyte and Thrombocyte Abnormalities<br />

With hypersegmentation, i.e. 4–5 segments/nucleus, the segmented<br />

granulocytes show all the indications <strong>of</strong> a maturation disorder. In addition,<br />

the reticulocyte count is increased (but it may also be normal), and<br />

the iron content is elevated or normal.<br />

Megaloblastic anemias show the same hematological picture whether<br />

they are caused by folic acid deficiency or by vitamin B12 deficiency.<br />

Table 26 lists possible causes. It should be emphasized that “genuine” pernicious<br />

anemia is less common than megaloblastic anemia due to vitamin<br />

B12 deficiency. In pernicious anemia a stomach biopsy shows atrophic<br />

gastritis and usually also serum antibodies to parietal cells and intrinsic<br />

factor.<br />

Among the causes <strong>of</strong> folic acid deficiency is chronic alcoholism (with<br />

insufficient dietary folic acid, impaired absorption, and elevated erythrocyte<br />

turnover). On the other hand, many alcoholics with normal vitamin<br />

B12 and folic acid levels develop severe hyperchromic anemia with a<br />

special bone marrow morphology, obviously with a pathomechanism <strong>of</strong><br />

its own (pyridoxine [B6] deficiency, among others).<br />

In megaloblastic anemia (Fig. 53) the cell density in the bone marrow is<br />

always remarkably high. Large to medium-sized blasts with round nuclei<br />

dominate the erythrocyte series. They are present in varying sizes, their<br />

chromatin is loosely arranged with a coarse “sandy” reticular structure,<br />

there are well-defined nucleoli, and the cytoplasm is very basophilic with<br />

a perinuclear lighter zone. These cells can be interpreted as proerythroblasts<br />

and macroblasts whose maturation has been disturbed. As these<br />

disturbed megaloblastic cells appear along a continuous spectrum from<br />

the less mature to the more mature, they are all referred to collectively as<br />

megaloblasts.<br />

In the granylocytic series, anomalies become obvious at the myelocyte<br />

stage; characteristic giant cells with loosely structured nuclei develop<br />

which may tend to be classified as myelocytes/stab cells, but which in fact<br />

probably are myelocytes in which the maturation process has been disturbed.<br />

As in peripheral blood smears, segmented granulocytes are <strong>of</strong>ten<br />

hypersegmented. Megakaryocytes also show hypersegmentation <strong>of</strong> their<br />

nuclei or many individual nuclei. Iron staining reveals increased number<br />

<strong>of</strong> iron-containing reticular cells and sideroblasts, and a few ring sideroblasts<br />

may develop. All these changes disappear after vitamin B12 supplementation,<br />

after just three days in the erythrocyte series and within<br />

one week in the granulocyte series. In the differential diagnosis, in relation<br />

to the causes listed in Table 26, the following should be highlighted: toxic<br />

alcohol damage (vacuolized proerythroblasts), hemolytic anemia (elevated<br />

reticulocyte count), myelodysplasia (for bone marrow morphology see<br />

Fig. 37, p. 109).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

In older patients, myelodysplastic syndrome should be the first<br />

item in the differential diagnosis <strong>of</strong> hyperchromic anemias<br />

b c<br />

Fig. 54 Myelodysplastic syndrome (MDS) as differential diagnosis in hyperchromic<br />

anemia. a Strongly basophilic stippling in the cytoplasm <strong>of</strong> a macrocyte<br />

(in myelodysplasia). b Myeloblast with hyperchromic erythrocyte as an example<br />

<strong>of</strong> a myelodysplastic blood sample in the differential diagnosis versus hyperchromic<br />

anemia. c A high proportion <strong>of</strong> reticulocytes speaks against megaloblastic<br />

anemia and for hemolysis (in this case with an absence <strong>of</strong> pyruvate kinase activity).<br />

d Bone marrow in myelodysplasia (type RAEB), with clinical hyperchromic<br />

anemia.<br />

155<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

d


156<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Erythrocyte Inclusions<br />

Erythrocytes normally show fairly homogeneous hemoglobinization after<br />

panoptic staining; only after supravital staining (p. 155) will the remains<br />

<strong>of</strong> their ribosomes show as substantia granul<strong>of</strong>ilamentosa in the reticulocytes.<br />

Under certain conditions during the preparation <strong>of</strong> erythrocytes,<br />

aggregates <strong>of</strong> ribosomal material may be seen as basophilic stippling, also<br />

visible after Giemsa staining (Fig. 55 a). This is normal in fetal and infant<br />

blood; in adults a small degree <strong>of</strong> basophilic stippling has nonspecific diagnostic<br />

value, like anisocytosis indicating only a nonspecific reactive<br />

process. Not until a large proportion <strong>of</strong> basophilic stippled erythrocytes is<br />

seen concomitantly with anemia does this phenomenon have diagnostic<br />

significance: thalassemia, lead poisoning, and sideroblastic anemia are all<br />

possibilities. Errant chromosomes from the mitotic spindle are sometimes<br />

found in normoblasts as small spheres with a diameter <strong>of</strong> about 1 µm,<br />

which have remained in the erythrocyte after expulsion <strong>of</strong> the nucleus and<br />

lie about as unevenly distributed Howell–Jolly bodies in the cytoplasm<br />

(Fig. 55 b, c). Normally, these cells are quickly sequestered by the spleen.<br />

Always after splenectomy, rarely also in hemolytic conditions and megaloblastic<br />

anemia, they are observed in erythrocytes at a rate <strong>of</strong> 1‰.<br />

These Howell-Jolly bodies are visible after normal panoptic staining.<br />

Supravital staining (see reticulocyte count, p. 11) may produce a few<br />

globular precipitates at the cell membrane, known as Heinz bodies. They<br />

are a rare phenomenon and may indicate the presence <strong>of</strong> unstable<br />

hemoglobins in rare familial or severe toxic hemolytic conditions (p. 144).<br />

An ellipsoid, eosinophil-violet stained, delicately structured ring in<br />

erythrocytes is called a Cabot ring (Fig. 55 d). It probably consists not, as its<br />

form might suggest, <strong>of</strong> remnants <strong>of</strong> the nuclear membrane, but <strong>of</strong> fibers<br />

from the mitotic spindle. Because these ring structures are sporadically<br />

seen in all severe anemias, no specific conclusions can be drawn from their<br />

presence, but in the absence <strong>of</strong> any other rationale for severe anemia they<br />

are compatible with the suspicion <strong>of</strong> an incipient idiopathic erythropoietic<br />

disorder (e.g., panmyelopathy or smoldering leukosis or<br />

leukemia).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

Small inclusions are usually a sign <strong>of</strong> nonspecific anomalies or<br />

artifacts<br />

d e<br />

f g<br />

Fig. 55 Erythrocyte inclusions. a Polychromatic erythrocyte with fine, dense basophilic<br />

stippling. b Erythrocyte with Howell–Jolly bodies (arrow) in addition to a<br />

lymphocyte (after splenectomy). c Erythrocyte with two Howell–Jolly bodies (arrow)<br />

alongside an orthochromatic erythroblast with basophilic stippling (thalassemia,<br />

in this case with functional asplenia). d Erythrocyte with a delicate Cabot<br />

ring (arrow) (here in a case <strong>of</strong> osteomyelosclerosis). e Thrombocyte layered onto<br />

an erythrocyte (arrow). f and g Fixation and staining artifacts.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

157<br />

b<br />

c


158<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Hematological Diagnosis <strong>of</strong> Malaria<br />

Various parasites may be found in the blood stream, e.g., trypanosomes<br />

and filariae. Among the parasitic diseases, probably only malaria is <strong>of</strong><br />

practical diagnostic relevance in the northern hemisphere, while at the<br />

same time malarial involvement <strong>of</strong> erythrocytes may confuse the interpretation<br />

<strong>of</strong> erythrocyte morphology. For these reasons, a knowledge <strong>of</strong><br />

the principal different morphological forms <strong>of</strong> malarial plasmodia is<br />

advisable.<br />

Recurrent fever and influenza-like symptoms after a stay in tropical regions<br />

suggest malaria. The diagnosis may be confirmed from normal<br />

blood smears or thick smears; in the latter the erythrocytes have been<br />

hemolyzed and the pathogens exposed. Depending on the stage in the life<br />

cycle <strong>of</strong> the plasmodia, a variety <strong>of</strong> morphologically completely different<br />

forms may be found in the erythrocytes, sometimes even next to each<br />

other. The different types <strong>of</strong> pathogens show subtle specific differences<br />

that, once the referring physician suspects malaria, are best left to the<br />

specialist in tropical medicine, who will determine which <strong>of</strong> the following<br />

is the causative organism: Plasmodium vivax (tertian malaria), Plasmodium<br />

falciparum (falciparum or malignant tertian malaria) and Plasmodium<br />

malariae (quartan malaria) (Table 27, Fig. 56). Most cases <strong>of</strong><br />

malaria are caused by P. vivax (42%) and P. falciparum (43%).<br />

The key morphological characteristics in all forms <strong>of</strong> malaria can be<br />

summarized as the following basic forms: The first developmental stage <strong>of</strong><br />

the pathogen, generally found in quite large erythrocytes, appears as small<br />

ring-shaped bodies with a central vacuole, called trophozoites (or the<br />

signet-ring stage) (Fig. 57). The point-like center is usually most noticeable,<br />

as it is reminiscent <strong>of</strong> a Howell–Jolly body, and only a very careful<br />

search for the delicate ring form will supply the diagnosis. Occasionally,<br />

there are several signet-ring entities in one erythrocyte. All invaded cells<br />

may show reddish stippling, known as malarial stippling or Schüffner’s<br />

dots. The pathogens keep dividing, progressively filling the vacuoles, and<br />

then develop into schizonts (Fig. 56), which soon fill the entire erythrocyte<br />

with an average <strong>of</strong> 10–15 nuclei. Some schizonts have brown-black pigment<br />

inclusions. Finally the schizonts disintegrate, the erythrocyte ruptures,<br />

and the host runs a fever. The separate parts (merozoites) then start a<br />

new invasion <strong>of</strong> erythrocytes.<br />

In parallel with asexual reproduction, merozoites develop into gamonts,<br />

which always remain mononuclear and can completely fill the<br />

erythrocyte with their stippled cell bodies. The large, dark blue-stained<br />

forms (macrogametes) are female cells (Fig. 57 d), the smaller, light bluestained<br />

forms (microgametes) are male cells. Macrogametes usually predominate.<br />

They continue their development in the stomach <strong>of</strong> the infected<br />

Anopheles mosquito.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Young<br />

trophozoite<br />

Intermediate<br />

trophozoite<br />

Mature<br />

schizont<br />

Mature<br />

macrogametocyte<br />

Mature<br />

microgametocyte<br />

Plasmodium<br />

falciparum<br />

Plasmodium<br />

vivax<br />

Plasmodium<br />

ovale<br />

Plasmodium<br />

malariae<br />

Fig. 56 Differential diagnosis <strong>of</strong> malaria plasmodia in a blood smear (from Kayser,<br />

F., et al., Medizinische Mikrobiologie (Medical Microbiology), Thieme, Stuttgart,<br />

1993).<br />

159<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


160<br />

Table 27 Parasitology <strong>of</strong> malaria and clinical characteristics (from Diesfeld, H., G.<br />

Krause: Praktische Tropenmedizin und Reisemedizin. Thieme, Stuttgart 1997)<br />

Disease Pathogen Exoerythrocytic<br />

phase<br />

incubation<br />

Falciparum<br />

malaria<br />

Tertian<br />

malaria<br />

Quartan<br />

malaria<br />

Erythrocyte and Thrombocyte Abnormalities<br />

The example shown here is <strong>of</strong> the erythrocytic phases <strong>of</strong> P. vivax, because<br />

tertian malaria is the most common form <strong>of</strong> malaria and best shows<br />

the basic morphological characteristics <strong>of</strong> a plasmodia infection.<br />

Plasmodium<br />

falciparum<br />

P. vivax<br />

P. ovale<br />

⎫<br />

⎪<br />

⎬<br />

⎪<br />

⎭<br />

7–15 days, does<br />

not form liver<br />

hypnozoites<br />

12-18 days,<br />

forms liver<br />

hypnozoites<br />

P. malariae 18-40 days,<br />

does not form<br />

liver hypnozoites<br />

Erythrocytic<br />

phase<br />

48 hours,<br />

recurring<br />

fever is rare<br />

48 hours<br />

Clinical characteristics<br />

Potentially lethal<br />

disease, widely<br />

therapy-resistant,<br />

there is usually no<br />

recurrence after<br />

recovery<br />

Benign form recurrences<br />

up to 2 years<br />

after infection<br />

Benign form recurrences<br />

up to 5 years<br />

after infection<br />

72 hours Benign form recurrences<br />

possible up<br />

to 30 years after<br />

infection<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

Conspicuous erythrocyte inclusions suggest malaria<br />

b c<br />

d e<br />

Fig. 57 Blood analysis in malaria. a Trophozoites in Plasmodium falciparum (falciparum<br />

or malignant tertian malaria) infection. Simple signet-ring form (1), double-invaded<br />

erythrocyte with basophilic stippling (2). Erythrocyte with basophilic<br />

stippling without plasmodium (3) and segmented neutrophilic granulocyte with<br />

toxic granulation (4). b and c Plasmodium falciparum: single invasion with delicate<br />

trophozoites (1), multiple invasion (2). d and e In the gametocyte stage <strong>of</strong> falciform<br />

malaria, the pathogens appear to reside outside the erythrocyte, but remnants<br />

<strong>of</strong> the erythrocyte membrane may be seen (arrow, e). For a systematic overview<br />

<strong>of</strong> malarial inclusions, see p. 159.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

161


162<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Polycythemia Vera (Erythremic<br />

Polycythemia) and Erythrocytosis<br />

Increases in erythrocytes, hemoglobin, and hematocrit above the normal<br />

range due to causes unrelated to hematopoiesis (i.e., the majority <strong>of</strong> cases)<br />

are referred to as secondary erythrocytosis or secondary polycythemia.<br />

However, it should be remembered that the “normal” range <strong>of</strong> values is<br />

quite wide, especially for men, in whom the normal range can be as much<br />

as 55% <strong>of</strong> the hematocrit!<br />

Table 28 Causes <strong>of</strong> secondary erythrocytosis<br />

Reduced O2 transport capacity – Carboxyhemoglobin formation in chronic<br />

smokers<br />

– High altitude hypoxia, COPD<br />

– Heart defect (right–left shunt)<br />

– Hypoventilation (e.g., obesity hypoventilation)<br />

Reduced O2 release from hemoglobin<br />

Renal hypoxia<br />

Autonomous erythropoietin biosynthesis – Renal carcinoma<br />

– Adenoma<br />

COPD = Chronic obstructive pulmonary disease<br />

– Congenital 2,3-diphosphoglycerate deficiency<br />

– Hydronephrosis, renal cyst<br />

– Renal artery stenosis<br />

An autonomous increase in erythropoiesis, i.e., polycythemia vera, represents<br />

a very different situation. Polycythemia vera—which one might<br />

call a “primary erythrocytosis“—is a malignant stem cell abnormality <strong>of</strong><br />

unknown origin and is a myeloproliferative syndrome (p. 112). Leukocyte<br />

counts (with increased basophils) and thrombocyte counts <strong>of</strong>ten rise<br />

simultaneously, and a transition to osteomyelosclerosis <strong>of</strong>ten occurs in<br />

the later stages.<br />

Revised Criteria for the Diagnosis <strong>of</strong> Polycythemia Vera (according to<br />

Pearson and Messinezy, 1996)<br />

— A1 elevated erythrocyte numbers<br />

— A2 absence <strong>of</strong> any trigger <strong>of</strong> secondary polycythemia<br />

— A3 palpable splenomegaly<br />

— A4 clonality test, e.g., PRV-1 marker (polycythemia rubra vera 1)<br />

— B1 thrombocytosis ( 400 10 9 /l)<br />

— B2 leukocytosis ( 10 10 9 /l)<br />

— B3 splenomegaly (sonogram)<br />

— B4 endogenous erythrocytic colonies<br />

Diagnosis in the presence <strong>of</strong>: A1 + A2 + A3/A4 or A1 + A2 + 2 B<br />

Bone marrow cytology shows increased erythropoiesis in both polycythemia<br />

vera and secondary polycythemia. Polycythemia vera is the<br />

more likely diagnosis when megakaryocytes, granulopoiesis, basophils,<br />

and eosinophils are also increased.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

d<br />

Bone marrow analysis contributes to the differential diagnosis<br />

between secondary erythrocytosis and polycythemia vera<br />

Fig. 58 Polycythemia vera and secondary erythrocytosis. a In reactive secondary<br />

erythrocytosis there is usually only an increase in erythropoiesis. b In polycythemia<br />

vera megakaryopoiesis (and <strong>of</strong>ten granulopoiesis) are also increased. c Bone<br />

marrow smear at low magnification in polycythemia vera, with a hyperlobulated<br />

megakaryocyte (arrow). d Bone marrow smear at low magnification in polycythemia<br />

vera, showing increased cell density and proliferation <strong>of</strong> megakaryocytes. e In<br />

polycythemia vera, iron staining shows no iron storage particles.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

163<br />

b<br />

e


164<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Thrombocyte Abnormalities<br />

EDTA in blood collection tubes (e.g., lavender top tubes) can lead to<br />

aggregation (pseudothrombocytopenia). A control using citrate as anticoagulant<br />

is required.<br />

The clinical sign <strong>of</strong> spontaneous bleeding in small areas <strong>of</strong> the skin and<br />

mucous membranes (petechial bleeding), which on injury diffuses out to<br />

form medium-sized subcutaneous ecchymoses, is grounds for suspicion<br />

<strong>of</strong> thrombocyte (or vascular) anomalies.<br />

Thrombocytopenia<br />

Thrombocytopenias Due to Increased Demand (High Turnover)<br />

A characteristic sign <strong>of</strong> this pathology can be that among the few thrombocytes<br />

seen in the blood smear, there is an increased presence <strong>of</strong> larger,<br />

i.e., less mature cell forms.<br />

The bone marrow in these thrombocytopenias shows raised (or at least<br />

normal) megakaryocyte counts. Here too, there may be an increased presence<br />

<strong>of</strong> less mature forms with one or two nuclei (Figs. 60, 61).<br />

Drug-induced immunothrombocytopenia As in drug-induced agranulocytosis,<br />

the process starts with an antibody response to a drug, or its metabolites,<br />

and binding <strong>of</strong> the antibody complex to thrombocytes. Rapid<br />

thrombocyte degradation by macrophages follows (Table 29). The most<br />

common triggers are analgesic/anti-inflammatory drugs and antibiotics.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Thrombocytes: increases, reductions, and anomalies may be<br />

seen<br />

b<br />

Fig. 59 Forms <strong>of</strong> thrombocytopenia. a This blood smear shows normal size and<br />

density <strong>of</strong> thrombocytes. b In this blood smear thrombocyte density is lower and<br />

size has increased, a feature typical <strong>of</strong> immunothrombocytopenia. continued <br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

165<br />

a


166<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Table 29 Most common triggers <strong>of</strong> drug-induced immunothrombocytopenia<br />

Analgesics<br />

Antibiotics<br />

Anticonvulsive drugs<br />

Arsenic, e. g., in water<br />

Quinidine<br />

Quinine<br />

Digitalis preparations<br />

Furosemide<br />

Gold salts<br />

Heparin!<br />

Isoniazid<br />

Methyldopa<br />

Spironolactone<br />

Tolbutamide<br />

“Idiopathic” immunothrombocytopenia. The name is largely historical,<br />

because <strong>of</strong>ten a trigger is found.<br />

➤ Postinfectious = acute form, usually occurs in children after rubella,<br />

mumps, or measles.<br />

➤ Essential thrombocytopenia, thrombocytopenic purpura (Werlh<strong>of</strong> disease)<br />

Thrombocyte antibodies develop without an identifiable trigger. This is<br />

thus a primary autoimmune disease specifically targeting thrombocytes.<br />

Secondary immunothrombocytopenias, e.g., in lupus erythematosus and<br />

other forms <strong>of</strong> immune vasculitis, lymphomas, and tuberculosis.<br />

Post-transfusion purpura occurs mostly in women about one week after a<br />

blood transfusion, <strong>of</strong>ten after earlier transfusions or pregnancy.<br />

Thrombocytopenia in microangiopathy. This group includes thrombotic–<br />

thrombocytopenic purpura (see p. 144) and disseminated intravascular<br />

coagulation.<br />

Thrombocytopenia in hypersplenism <strong>of</strong> whatever etiology.<br />

Fig. 59 Continued. c Pseudothrombocytopenia. The thrombocytes are not lying<br />

free and scattered around, but agglutinated together, leading to a reading <strong>of</strong><br />

thrombocytopenia from the automated blood analyzer. d Giant thrombocyte (as<br />

large as an erythrocyte) in thrombocytopenia. Döhle-type bluish inclusion (arrow)<br />

in the normally granulated segmented neutrophilic granulocyte: May-Hegglin<br />

anomaly.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Thrombocytes: increases, reductions, and anomalies may be<br />

seen<br />

c d<br />

Fig. 59 e Large thrombocyte (1) in thrombocytopenia. Thrombocyte-like fragments<br />

from destroyed granulocytes (cytoplasmic fragments) (2), which have the<br />

same structure and staining characteristics as the cytoplasm <strong>of</strong> band granulocytes<br />

(3). Clinical status <strong>of</strong> sepsis with disseminated intravascular coagulation. In automated<br />

counters cytoplasmic fragments are included in the thrombocyte fraction.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

167<br />

e


168<br />

Thrombocytopenias Due to Reduced Cell Production<br />

In this condition, blood contains few, usually small, pyknotic (“old”)<br />

thrombocytes. Only a very few megakaryocytes are found in the bone<br />

marrow, and these have a normal appearance.<br />

Causes<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Chronic alcoholism. There may be some overlap with increased turnover<br />

and folic acid deficiency.<br />

Chemical and radiological noxae. Cytostatics naturally lead directly to a<br />

dose-dependent reduction in megakaryocyte counts. A large therapeutic<br />

radiation burden in the area <strong>of</strong> blood-producing bone marrow has the<br />

same result, which may persist for many months.<br />

Virus infections. Measles, mononucleosis (Epstein–Barr virus, cytomegalovirus),<br />

rubella, and influenza may (usually in children) trigger thrombocytopenias<br />

<strong>of</strong> various types. In these cases, the virus affects the megakaryocytes<br />

directly. However, antibodies to thrombocytes may also arise<br />

in the course <strong>of</strong> these infections (p. 166), so that the pathomechanism <strong>of</strong><br />

the parainfectious thrombocytopenias described by Werlh<strong>of</strong> in children<br />

may lie in impaired production and/or increased degradation <strong>of</strong> thrombocytes.<br />

Neoplastic and aplastic bone marrow diseases. All neoplasms <strong>of</strong> the bone<br />

marrow cell series (e.g., leukemia, lymphoma, and plasmacytoma), together<br />

with their precursor forms (e.g., myelodysplasia), lead to progressive<br />

thrombocytopenia, as do panmyelophthisis and bone marrow infiltration<br />

by metastases from solid tumors.<br />

Vitamin deficiency. Folic acid and vitamin B12 deficiencies from various<br />

causes (p. 152) also affect the rapidly proliferating megakaryocytes. In<br />

these cases, thrombocytopenia is <strong>of</strong>ten present before anemia and<br />

leukocytopenia in circulating blood, while the bone marrow shows<br />

copious megakaryocytes that have been blocked from maturation.<br />

Constitutional diseases. An amegakaryocytic thrombocytopenia without<br />

any <strong>of</strong> the above causes is rare. It is seen in children with congenital radial<br />

aplasia; in adults it tends usually to be an early sign <strong>of</strong> leukemia,<br />

myelodysplastic syndrome, or aplastic anemia.<br />

Wiscott-Aldrich syndrome (thrombocytopenia, immune deficiency, and<br />

eczema) is an X-chromosomal recessive disease in boys and presents with<br />

thrombocytopenia with ineffective megakaryopoiesis.<br />

The May-Hegglin anomaly (dominant hereditary transmission) is<br />

characterized by thrombocytopenia with giant thrombocytes and<br />

granulocyte inclusions, which resemble Döhle bodies (endoplasmatic reticulum<br />

aggregates).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Variant forms <strong>of</strong> thrombocyte and megakaryocyte morphology<br />

in the bone marrow are diagnostic aids in thrombocytopenia<br />

e<br />

f<br />

Fig. 60 Morphology <strong>of</strong> thrombocytes and megakaryocytes. a Bone marrow in<br />

thrombocytopenia due to increased turnover (e.g., immunothrombocytopenia).<br />

Mononuclear “young” megakaryocytes clearly budding a thrombocyte (irregular,<br />

cloudy cytoplasm structure). b In thrombocytopenia against a background <strong>of</strong><br />

myelodysplasia, the bone marrow shows various megakaryocyte anomalies: here,<br />

too small a nucleus surrounded by too wide cytoplasm. c–f In myelodysplasia<br />

(c and d) and acute myeloid leukemia (e and f), bizarre anomalous thrombocyte<br />

shapes (arrows) may occasionally be found.<br />

169<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

b<br />

d


170<br />

Erythrocyte and Thrombocyte Abnormalities<br />

Thrombocytosis (Including<br />

Essential Thrombocythemia)<br />

Reactive. Thrombocytosis in the form <strong>of</strong> a constant elevation <strong>of</strong> thrombocyte<br />

counts above an upper normal range <strong>of</strong> 300 000–450 000/µl, may be<br />

reactive, i.e., may appear in response to various tumors (particularly<br />

bronchial carcinoma), chronic inflammation (particularly ulcerative colitis,<br />

primary chronic polyarthritis), bleeding, or iron deficiency. The<br />

pathology <strong>of</strong> this form is not known.<br />

Essential Thrombocythemia<br />

Essential thrombocythemia, by contrast, is a myeloproliferative disease<br />

(see p. 114) in which the main feature <strong>of</strong> increased thrombocytes is accompanied<br />

by other signs <strong>of</strong> this group <strong>of</strong> diseases that may vary in severity,<br />

such as leukocytosis and an enlarged spleen. Severe thrombocythemia<br />

may also be seen in osteomyelosclerosis, polycythemia vera, and chronic<br />

myeloid leukemia, and for this reason the following specific diagnostic criteria<br />

have been suggested:<br />

Diagnostic criteria for essential thrombocytopenia (according to<br />

Murphy et al.)<br />

➤ Thrombocytes 600 10 9 /l (with control)<br />

➤ Normal erythrocyte mass or Hb 18.5 g/dl , 16.5 g/dl <br />

➤ No significant bone marrow fibrosis<br />

➤ No splenomegaly<br />

➤ No leukoerythroblastic CBC<br />

➤ Absence <strong>of</strong> morphological or cytogenetic criteria <strong>of</strong> myelodysplasia<br />

➤ Secondary thrombocytosis (iron deficiency, inflammation, neoplasia,<br />

trauma, etc.) excluded<br />

Large thrombocytes are found in the peripheral blood smear. However,<br />

these also occur in polycythemia vera and osteomyelosclerosis.<br />

Bone marrow cytology will show markedly elevated megakaryocyte<br />

counts, with the cells <strong>of</strong>ten forming clusters and <strong>of</strong>ten with hypersegmented<br />

nuclei.<br />

Fig. 61 Essential thrombocythemia. a Increased thrombocyte density and marked<br />

anisocytosis in essential thrombocythemia. b Large thrombocytes (1) and a<br />

micro(mega)karyocyte nucleus (2) in essential thrombocythemia. Micro(mega)karyocytes<br />

are characterized by a small, very dense and <strong>of</strong>ten lobed nucleus<br />

with narrow, uneven cytoplasm, the processes <strong>of</strong> which correspond to thrombocytes<br />

(arrow).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

Thrombocyte proliferation with large megakaryocytes: essential<br />

thrombocythemia, a chronic myeloproliferative disease<br />

Fig. 61 c and d Bone marrow cytology in essential thrombocythemia: there is a<br />

striking abundance <strong>of</strong> very large, hyperlobulated megakaryocytes (c); such megakaryocytes<br />

may also be seen in polycythemia vera. d Size comparison with basophilic<br />

erythroblasts (arrow). The cloudy cytoplasm <strong>of</strong> the megakaryocyte is typical<br />

<strong>of</strong> effective thrombocyte production.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

171<br />

b<br />

d


172<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Cytology <strong>of</strong> Organ Biopsies<br />

and Exudates*<br />

* Special thanks to Dr. T. Binder, Wuppertal,<br />

for the generous gift <strong>of</strong> several preparations.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


174<br />

Cytology <strong>of</strong> Organ Biopsies and Exudates<br />

In this guide to morphology, only a basic indication can be given <strong>of</strong> the<br />

materials that may be drawn upon for a cytological diagnosis and what<br />

basic kinds <strong>of</strong> information cytology is able to give.<br />

For specialized cytological organ diagnostics, the reader should refer to<br />

a suitable cytology atlas. Often appropriately prepared samples are <strong>of</strong>ten<br />

sent away to a hematological–cytological or a pathoanatomic laboratory<br />

for analysis. Thus, the images in this chapter are intended particularly to<br />

help the clinician understand the interpretation <strong>of</strong> samples that he or she<br />

has not investigated in person.<br />

In principle, all parenchymatous organs can be accessed for material for<br />

cytological analysis. Of particular importance are thyroid biopsy (especially<br />

in the region <strong>of</strong> scintigraphically “cold” nodules), liver and spleen<br />

biopsy (under laparascopic guidance) in the region <strong>of</strong> lumps lying close to<br />

the surface, and breast and prostate biopsy. Again, the cytological analysis<br />

is usually made by a specialist cytologist or pathologist.<br />

Lymph node cytology, effusion cytology (pleura, ascites), cerebrospinal<br />

fluid cytology, and bronchial lavage are usually the responsibility <strong>of</strong> the<br />

internist with a special interest in morphology and are closely related to<br />

hemato-oncology.<br />

Lymph Node Cytology<br />

The diagnosis <strong>of</strong> enlarged lymph nodes receives special attention here because<br />

lymph nodes are as important as bone marrow for hematopoiesis.<br />

While in most instances abnormalities in the bone marrow cell series can<br />

be detected from the peripheral blood, this is very rarely the case for lymphomas.<br />

For this reason, lymph node cytology, a relatively simple and<br />

well-tolerated technique (p. 24), is critically important for the guidance it<br />

can give about the cause <strong>of</strong> enlarged lymph nodes. Figure 62 <strong>of</strong>fers a diagnostic<br />

flow chart.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Anamnesis – sudden fast swelling<br />

slow onset, unclear<br />

symptoms: subfebrile,<br />

night sweat, weight<br />

loss<br />

Findings – pressure pain<br />

indolent <strong>of</strong>ten in one<br />

Blood<br />

analysis<br />

Serology/ – mononucleosis test<br />

immunology – toxoplasmosis<br />

– rubella<br />

– syphilis<br />

– tuberculin skin test<br />

or Search<br />

for disease<br />

focus<br />

– possible onset in youth<br />

– possible contact<br />

with animals<br />

– possible fever<br />

– focal, or distributed<br />

over several<br />

locations<br />

– relative lymphocytosis<br />

with<br />

stimulated forms<br />

– e.g. teeth<br />

– sinuses<br />

– infections <strong>of</strong><br />

the genitalia<br />

– if findings are<br />

negative or unsuccessful<br />

therapy<br />

after 1 week<br />

considered reactive if<br />

the lymph node swelling<br />

does not recede after<br />

2 weeks or the cause<br />

is found<br />

D<br />

D<br />

D<br />

attempt to clarify<br />

the diagnosis after histology<br />

Lymph node<br />

biopsy<br />

Lymph Node Cytology<br />

possibly<br />

location, possibly<br />

localization <strong>of</strong><br />

primary tumor<br />

175<br />

specific cell presentation<br />

(e.g. CLL, immunocytoma,<br />

ALL and others)<br />

or unspecific signs<br />

suspicion <strong>of</strong> tumor or<br />

malignant lymphoma<br />

D<br />

diagnosis based<br />

on histology<br />

Fig. 62 Diagnostic flow chart for cases <strong>of</strong> lymph node enlargement. (D) Diagnosis.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


176<br />

Cytology <strong>of</strong> Organ Biopsies and Exudates<br />

Reactive Lymph Node Hyperplasia and<br />

Lymphogranulomatosis (Hodgkin Disease)<br />

Reactive lymph node hyperplasia <strong>of</strong> whatever etiology is characterized by a<br />

confused mixture <strong>of</strong> small, middle-sized, and large lymphocytes. When<br />

the latter have a nuclear diameter at least three times the size <strong>of</strong> the predominating<br />

small lymphocytes and have a fair width <strong>of</strong> basophilic cytoplasm,<br />

they are called immunoblasts (lymphoblasts). Cells with deeply<br />

basophilic, eccentric cytoplasm and dense nuclei are called plasmablasts,<br />

and cells with a narrow cytoplasmic seam are centroblasts. Lymphocytes<br />

can also to varying degrees show a tendency to appear as plasma cells, e.g.,<br />

as plasmacytoid lymphocytes with a relatively wide seam <strong>of</strong> cytoplasm.<br />

Monocytes and phagocytic macrophages are also seen (Fig. 63).<br />

Table 30 (p. 178) summarizes the different forms <strong>of</strong> reactive lymphadenitis.<br />

The basic cytological findings in all <strong>of</strong> them is always a complete<br />

mixture <strong>of</strong> small to very large lymphocytes. Occasionally more specific<br />

findings may indicate the possibility <strong>of</strong> mononucleosis (increased immature<br />

monocytes) or toxoplasmosis (plasmablasts, phagocytic macrophages,<br />

and possibly epithelioid cells).<br />

Any enlargement <strong>of</strong> the lymph nodes that persists for more than two<br />

weeks should be subjected to histological analysis unless the history, clinical<br />

findings, serology, or CBC <strong>of</strong>fer an explanation.<br />

At first sight, the confusion visible in the cytological findings <strong>of</strong> lymphogranulomatosis<br />

(Hodgkin disease) is reminiscent <strong>of</strong> the picture in reactive<br />

hyperplasia (something which may be important for an understanding <strong>of</strong><br />

the pathology <strong>of</strong> this disease compared with other malignant neoplasms).<br />

However, some cells elements show signs <strong>of</strong> a strong immunological<br />

“over-reaction” in which large, immunoblast-like cells form with welldeveloped<br />

nucleoli (Hodgkin cells). Sporadically, some <strong>of</strong> these cells are<br />

found to be multinucleated (Reed–Sternberg giant cells); infiltrations <strong>of</strong><br />

eosinophils and plasma cells may also be found. Findings <strong>of</strong> this type always<br />

require histological analysis, which can distinguish between four<br />

prognostically relevant histological subtypes. In addition to this, the very<br />

lack <strong>of</strong> a clear demarcation between Hodgkin disease and reactive conditions<br />

is reason enough to conduct a histological study <strong>of</strong> every lymph node<br />

that appears reactive if does not regress completely within two weeks.<br />

In cases <strong>of</strong> histologically verified Hodgkin disease, cytological analysis<br />

is especially useful in the assessment <strong>of</strong> new lymphomas after therapy.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

Reactive lymph node hyperplasia and lymphogranulomatosis<br />

(Hodgkin disease): a polymorphous mixture <strong>of</strong> cells<br />

b c<br />

Fig. 63 Reactive lymph node hyperplasia and lymphogranulomatosis. a Lymph<br />

node cytology in severe reactive hyperplasia. Large blastic cells alongside small<br />

lymphocytes (if it fails to regress, histological analysis is required). b Hodgkin<br />

disease: a giant mononuclear cell with a large nucleolus (arrow) and wide cytoplasmic<br />

layer (Hodgkin cell), surrounded by small and medium-sized lymphocytes.<br />

c Hodgkin disease: giant binuclear cell (Reed–Sternberg giant cell).<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

177


Table 30 Sequence <strong>of</strong> steps in the diagnosis <strong>of</strong> reactive lymphoma<br />

178<br />

Anamnesis Symptoms Tentative diagnosis Diagnostic studies Cytology Histology<br />

() Non-specific<br />

lymphadenitis<br />

Search for disease<br />

focus, non-specific<br />

changes in CBC and<br />

ESR<br />

Localized, painful Perifocal lymphadenitis<br />

Rapid progression<br />

(fever)<br />

Cytology <strong>of</strong> Organ Biopsies and Exudates<br />

() Adenitis with<br />

histiocytosis<br />

Mononucleosis Pfeiffer cells in the<br />

blood analysis, EBV<br />

serology (1)<br />

Diffuse, painfult;<br />

angina, possibly<br />

spleen<br />

Rapid progression,<br />

fever,<br />

sore throat<br />

Rubella Plasma cells in the<br />

blood,<br />

rubella-AHT (2)<br />

Most children Nuchal lymph<br />

nodes, later<br />

exanthema<br />

Epithelioid cell<br />

lymphadenitis<br />

Diffuse Toxoplasmosis Serology (3) () With<br />

epithelioid cells and<br />

macrophages<br />

Ingestion <strong>of</strong> raw<br />

meat<br />

Contact with cats<br />

Complement fixation<br />

reaction (adenoviruses,<br />

less commonlycoxsackieviruses)<br />

(4)<br />

Common viruses,<br />

specific adenoviruses<br />

Local<br />

Neck area<br />

Pharyngitis<br />

( conjunctivitis)<br />

() Epithelioid<br />

cells, Langhans<br />

giant cells<br />

Tuberculosis Thorax, local primary<br />

infections, skin test,<br />

pathogen in biopsy<br />

aspirate, possibly PCR<br />

Inflamed lymph<br />

nodes, possibly<br />

fistulae<br />

Slow growth,<br />

general malaise<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

Epithelioid cells Epithelioid cell<br />

fibrosis<br />

Thorax, tuberculin<br />

test usually negative,<br />

ACE<br />

Sarcoidosis (Boeck<br />

disease)<br />

Slow growth Hard; possibly skin<br />

infiltrations


() It may be possible<br />

to determine the<br />

pathogen from the<br />

biopsy<br />

Listeriosis Agglutination test,<br />

complement fixation<br />

reaction<br />

Tonsillitis, neck<br />

lymph nodes<br />

Contact with<br />

animals<br />

Fever, spleen Brucellosis In case <strong>of</strong> fever:<br />

pathogen in blood,<br />

serology<br />

Contact with<br />

animals<br />

Milk intake<br />

Perforating<br />

lymphadenitis<br />

Epithelioid cells,<br />

giant cells<br />

Leukocytosis, lymphocytosis,complement<br />

fixation reaction<br />

Local primary lesion Cat-scratch disease<br />

Open wound,<br />

possibly from a<br />

cat<br />

Agglutination test<br />

Local primary lesion Tularemia (rabbit<br />

fever)<br />

Contact with<br />

wildlife<br />

Therapeutic<br />

excision<br />

Actinomycosis Leukocytosis, left shift “Gland” tissue in<br />

the biopsy material<br />

Inflamed hard infiltrates,<br />

possibly<br />

fistulae<br />

Little sense <strong>of</strong><br />

illness<br />

Lymph Node Cytology<br />

Collagen disease Antinuclear factor<br />

(PCP, LE) Felty syndrome,<br />

Still disease<br />

Branchial cyst Epithelial cells,<br />

macrophages, and<br />

granulocytes<br />

Joints, spleen,<br />

possibly kidney<br />

Symptomatic<br />

joints<br />

Therapeutic<br />

excision<br />

No symptoms Submandibular<br />

swelling, no irritation<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

() = Optional step; = usually diagnostic step, if there is no arrow, the diagnosis can be made on the basis <strong>of</strong> preceding steps. (1) = Positive from day 5;<br />

(2) = 1–4 days after exanthema, may be as much as 1 month; (3) = from week 4; (4) = from day 10.<br />

179


180<br />

Cytology <strong>of</strong> Organ Biopsies and Exudates<br />

Sarcoidosis and Tuberculosis<br />

The material <strong>of</strong> cell biopsies taken from indolent, nonirritated enlarged<br />

lymph nodes in the neck or axilla that have developed with little in the<br />

way <strong>of</strong> clinical symptoms, or from subcutaneous infiltration in various regions,<br />

can be quite homogeneous. With their thin, very long, ovoid nucleus<br />

(four to five times the size <strong>of</strong> lymphocytes), delicate reticular chromatin<br />

structure, and extensive layer <strong>of</strong> cytoplasm that may occasionally<br />

appear confluent with that <strong>of</strong> other cells, they are reminiscent <strong>of</strong> the<br />

epithelial cells that line the body’s internal cavities and are therefore<br />

called epithelioid cells. They are known to be the tissue form <strong>of</strong> transformed<br />

monocytes, and are found in increased numbers in all chronic inflammatory<br />

processes—especially toxoplasmosis, autoimmune diseases,<br />

and foreign-body reactions—and also in the neighborhood and drainage<br />

areas <strong>of</strong> tumors. They exclusively dominate the cytological picture in a<br />

particular form <strong>of</strong> chronic “inflammation,” sarcoidosis (Boeck disease). A<br />

typical finding almost always encountered at the pulmonary hilus combined<br />

with a negative tuberculin test will all but confirm this diagnosis.<br />

The appearance <strong>of</strong> a few multinuclear cells (Langhans giant cells) may<br />

allow confusion with tuberculosis, but clinical findings and a tuberculin<br />

skin test will usually make the diagnosis clear.<br />

Rapidly developing, usually hard, pressure-sensitive neck lymph nodes,<br />

seemingly connected with each other with some fluctuant zones and external<br />

inflammatory redness, suggest the now rare scr<strong>of</strong>ulous form <strong>of</strong><br />

tuberculosis. A highly positive tuberculin skin test also suggests this diagnosis.<br />

If any remaining doubts cannot be dispelled clinically, a very-fineneedle<br />

lymph node biopsy may be performed, but only if the skin shows<br />

noninflammatory, pale discoloration.<br />

The harvested material can show the potency <strong>of</strong> the tissue-bound<br />

forms <strong>of</strong> cells in the monocyte/macrophage series. In addition to mononuclear<br />

epithelioid cells, there are giant cell conglomerates made up <strong>of</strong><br />

polynuclear epithelioid cells in enormous syncytia with 10, 20, or more<br />

nuclei. These are called Langhans giant cells. In scr<strong>of</strong>uloderma (tuberculosis<br />

colliquativa), there are also lymphocytic and granulocytic cells in the<br />

process <strong>of</strong> degradation, which are absent in purely productive tuberculous<br />

lymphadenitis.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Epithelioid cells dominate the lymph node biopsy: Boeck disease<br />

or tuberculosis<br />

Fig. 64 Boeck disease and tuberculosis. a Lymph node cytology in Boeck disease:<br />

a special form <strong>of</strong> reactive cell pattern with (<strong>of</strong>ten predominating) islands<br />

and trains <strong>of</strong> epithelioid cells (arrow), which have ovoid nuclei with delicate chromatin<br />

structure and a wide, smoke-gray layer <strong>of</strong> cytoplasm. b Lymph node cytology<br />

in tuberculous lymphadenitis: in addition to lymphocytes and a few epithelial<br />

cells (1), enormous syncytes <strong>of</strong> epithelioid cell nuclei within one cytoplasm (arrow)<br />

may be encountered: the Langhans giant cell.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

181<br />

a<br />

b


182<br />

Cytology <strong>of</strong> Organ Biopsies and Exudates<br />

Non-Hodgkin Lymphoma<br />

Since the CBC is the first step in any lymph node diagnosis, lymph node biopsy<br />

is unnecessary in many cases <strong>of</strong> non-Hodgkin lymphoma (p. 70), because<br />

the most common form <strong>of</strong> this group <strong>of</strong> diseases, chronic lymphadenosis,<br />

can always be diagnosed on the basis <strong>of</strong> the leukemic findings <strong>of</strong> the<br />

CBC.<br />

However, when enlarged lymph nodes are found in one or more regions<br />

without symptoms <strong>of</strong> reactive disease, and the blood analysis fails to show<br />

signs <strong>of</strong> leukemia, lymph node biopsy is indicated.<br />

The relatively monotonous lymph node cytology in non-Hodgkin lymphomas<br />

and tumor metastases mean that histological differentiation is<br />

required.<br />

In contrast to Hodgkin disease, with its conspicuous giant cell forms<br />

(p. 177), non-Hodgkin lymphomas display a monotonous picture without<br />

any signs <strong>of</strong> a reactive process (p. 70). Clinically, it is enough to distinguish<br />

between small cell forms (which have a relatively good prognosis) and<br />

large cell forms (which have a poorer prognosis) to begin with. For a more<br />

detailed classification, see page 70 f.<br />

Histological analysis may be omitted only when its final results would<br />

not be expected to add to the intermediate cytological findings in terms <strong>of</strong><br />

consequences for treatment.<br />

Metastases <strong>of</strong> Solid Tumors in Lymph Nodes or<br />

Subcutaneous Tissue<br />

When hard nodules are found that are circumscribed in location, biopsy<br />

shows aggregates <strong>of</strong> polymorphous cells with mostly undifferentiated nuclei<br />

and a coarse reticular structure <strong>of</strong> the chromatin (perhaps with welldefined<br />

nucleoli or nuclear vacuoles), and the lymphatic cells cannot be<br />

classified, there is urgent suspicion <strong>of</strong> metastasis from a malignant solid<br />

tumor, i.e. from a carcinoma in a variety <strong>of</strong> possible locations or a s<strong>of</strong>t<br />

tissue sarcoma.<br />

As a rule, the next step is the search for a possible primary tumor. If this is<br />

found, lymph node resection becomes unnecessary.<br />

If no primary tumor is found, lymph node histology is indicated. The histological<br />

findings can provide certain clues about the etiology and also helps<br />

in the difficult differential diagnosis versus blastic non-Hodgkin lymphoma.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

In cases <strong>of</strong> non-Hodgkin lymphoma and tumor metastases, a<br />

tentative diagnosis is possible on the basis <strong>of</strong> the lymph node<br />

cytology<br />

d<br />

e<br />

Fig. 65 Non-Hodgkin lymphoma and tumor metastases. a Lymph node cytology<br />

showing small cells with relatively wide cytoplasm (arrow 1) in addition to lymphocytes.<br />

There are scattered blasts with wide cytoplasm (arrow 2): lymphoplasmacytic<br />

immunocytoma. b Lymph node cytology showing exclusively large blastoid<br />

cells with a large central nucleolus (arrow). This usually indicates large-cell non-<br />

Hodgkin lymphoma (in this case immunoblastic). c–e Metastatic disease from:<br />

c uterine carcinoma, d small-cell bronchial carcinoma, and e leiomyosarcoma.<br />

183<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

b


184<br />

Cytology <strong>of</strong> Organ Biopsies and Exudates<br />

Branchial Cysts and Bronchoalveolar<br />

Lavage<br />

Branchial Cysts<br />

A (usually unilateral) swollen neck nodule below the mandibular angle<br />

that feels firm to pressure, but is without external signs <strong>of</strong> inflammation,<br />

should suggest the presence <strong>of</strong> a branchial cyst. Surprisingly, aspiration<br />

usually produces a brownish-yellow liquid. In addition to partially cytolysed<br />

granulocytes and lymphocytes (cell detritus), a smear <strong>of</strong> this liquid,<br />

or the centrifuged precipitate, shows cells with small central nuclei and<br />

wide light cell centers which are identical to epithelial cells from the floor<br />

<strong>of</strong> the mouth. Biopsies from a s<strong>of</strong>t swelling around the larynx show the<br />

same picture; in this case it is a retention cyst from another developmental<br />

remnant, the ductus thyroglossus.<br />

Cytology <strong>of</strong> the Respiratory System,<br />

Especially Bronchoalveolar Lavage<br />

Through the development <strong>of</strong> patient-friendly endoscopic techniques, diagnostic<br />

lavage (with 10–30 ml physiological saline solution) and its cytological<br />

workup are now in widespread use. This method is briefly mentioned<br />

here because <strong>of</strong> its broad interest for all medical pr<strong>of</strong>essionals with<br />

an interest in morphology; the interested reader is referred to the<br />

specialist literature (e.g. Costabel, 1994) for further information. Table 31<br />

lists the most important indications for bronchoalveolar lavage.<br />

Table 31 Clinical indications for bronchoalveolar lavage (according to Costabel<br />

1994)<br />

Interstitial infiltrates Alveolar infiltrates Pulmonary infiltrates in<br />

patients with immune<br />

deficiency<br />

Sarcoidosis (Boeck disease)<br />

Exogenous allergic alveolitis<br />

Drug-induced alveolitis<br />

Idiopathic pulmonary fibrosis<br />

Collagen disease<br />

Histiocytosis X<br />

Pneumoconioses<br />

Lymphangiosis carcinomatosa<br />

Pneumonia<br />

Alveolar hemorrhage<br />

Alveolar proteinosis<br />

Eosinophilic pneumonia<br />

Obliterating bronchiolitis<br />

HIV Infection<br />

Treatment with cytostatic<br />

agents<br />

Radiation sickness<br />

Immunosuppressive therapy<br />

Organ transplant<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

b<br />

Accessible cysts (e.g., branchial cysts) should be aspirated. Bronchial<br />

lavage is a cytological new discipline<br />

c d<br />

Fig. 66 Cyst biopsy and bronchoalveolar lavage. a Cytology <strong>of</strong> a lateral neck cyst:<br />

no lymphatic tissue, but epithelial cells from the floor <strong>of</strong> the mouth. b Normal<br />

ciliated epithelial cells with typical cytoplasmic processes. c Tumor cell conglomeration<br />

in small-cell bronchial carcinoma: conglomeration is typical <strong>of</strong> tumor cells.<br />

d Bronchoalveolar lavage in purulent bronchitis: a macrophage with pigment<br />

inclusion (arrow) is surrounded by segmented neutrophilic granulocytes.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

185


186<br />

Cytology <strong>of</strong> Organ Biopsies and Exudates<br />

Cytology <strong>of</strong> Pleural Effusions and Ascites<br />

Pleural effusions always require cytological diagnostic procedures unless<br />

they are secondary to a known disease, such as cardiac insufficiency or<br />

pneumonia, and recede on treatment <strong>of</strong> the primary disease.<br />

Pleura aspirates can be classified as exudates or transudates (the latter<br />

usually caused by hydrodynamic stasis). The specific density (measured<br />

with a simple areometer) <strong>of</strong> transudates, which are protein-poor, is between<br />

1008 and 1015 g/l, while for exudates it is greater than 1018 g/l.<br />

Cytological preparation may be done by gentle centrifugation <strong>of</strong> the<br />

aspirate (10 minutes at 300–500 rpm), which should be as fresh as<br />

possible; the supernatant is decanted and the sediment suspended in the<br />

residual fluid, which will collect on the bottom <strong>of</strong> the centrifuge tube.<br />

Nowadays, however, this procedure has been replaced by cytocentrifugation.<br />

Effusions that are noticeably rich in eosinophilic granulocytes should<br />

raise the suspicion <strong>of</strong> Hodgkin disease, generalized reaction to the presence<br />

<strong>of</strong> a tumor, or an allergic or autoimmune disorder. Purely lymphatic<br />

effusions are particularly suggestive <strong>of</strong> tuberculosis. In addition, all transudates<br />

and exudates contain various numbers <strong>of</strong> endothelial cells (particularly<br />

high in cases <strong>of</strong> bacterial pleuritis) that have been sloughed <strong>of</strong>f from<br />

the pleural lining.<br />

Any cell elements that do not fulfill the above criteria should be regarded<br />

as suspect for neoplastic transformation, especially if they occur in<br />

aggregates. Characteristics that in general terms support such a suspicion<br />

include extended size polymorphy, coarse chromatin structure, welldefined<br />

nucleoli, occasional polynucleated cells, nuclear and plasma<br />

vacuoles, and deep cytoplasmic basophilia. For practical reasons, special<br />

diagnostic procedures should always be initiated in these situations.<br />

What was said above in relation to the cell composition <strong>of</strong> pleural effusions<br />

also holds for ascites. Here too, the specific density may be determined<br />

and the Rivalta test to distinguish exudate from transudate carried<br />

out. Inflammatory exudates usually have a higher cell content; a strong<br />

predominance <strong>of</strong> lymphocytes may indicate tuberculosis. Like the pleura,<br />

the peritoneum is lined by phagocytotic endothelial cells which slough <strong>of</strong>f<br />

into the ascitic fluid and, depending on the extent <strong>of</strong> the fluid, may produce<br />

a polymorphous overall picture analogous to that <strong>of</strong> the pleural endothelial<br />

cells. It is not always easy to distinguish between such endothelial<br />

cells and malignant tumor metastases. However, the latter usually<br />

occur not alone but in coherent cell aggregates (“floating<br />

metastases”), the various individual elements <strong>of</strong> which typically show a<br />

coarse chromatin structure, wide variation in size, well-defined nucleoli,<br />

and deeply basophilic cytoplasm.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


c<br />

Tumor cells can be identified in pleural and ascites smears<br />

a b<br />

Fig. 67 Pleural effusion and ascites. a Pleural cytology, nonspecific exudate: dormant<br />

mesothelial cell (or serosal cover cell) (1), phagocytic macrophage with vacuoles<br />

(2), and monocytes (3), in addition to segmented neutrophilic granulocytes<br />

(4). b Cell composition in a pleural aspirate (prepared using a cytocentrifuge): variable<br />

cells, whose similarity to cells in acute leukemia should be established by cytochemistry<br />

and marker analysis: lymphoblastic lymphoma. c Ascites with tumor<br />

cell conglomerate, surrounded with granulocytes and monocytes, in this case <strong>of</strong><br />

ovarian carcinoma. d Ascites cytology with an island <strong>of</strong> tumor cells. This kind <strong>of</strong><br />

conglomeration is typical <strong>of</strong> tumor cells.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

187<br />

d


188<br />

Cytology <strong>of</strong> Organ Biopsies and Exudate<br />

Cytology <strong>of</strong> Cerebrospinal Fluid<br />

The first step in all hemato-oncological and neurological diagnostic<br />

assessments <strong>of</strong> cerebrospinal fluid is the quantitative and qualitative<br />

analysis <strong>of</strong> the cell composition (Table 32).<br />

Table 32 Emergency diagnostics <strong>of</strong> the liquor (according to Felgenhauer in Thomas 1998)<br />

➤ Pandy’s reaction<br />

➤ Cell count (Fuchs-Rosenthal chamber)<br />

➤ Smear (or cytocentrifuge preparation)<br />

– to analyze the cell differentiation and<br />

– to search for bacteria and roughly determine their types and prevalence<br />

➤ Gram stain<br />

➤ An additional determination <strong>of</strong> bacterial antigens may be done<br />

Using advanced cell diagnostic methods, lymphocyte subpopulations<br />

can be identified by immunocytology and marker analysis and cytogenetic<br />

tests carried out on tumor cells.<br />

Prevalence <strong>of</strong> neutrophilic granulocytes with strong pleocytosis suggests<br />

bacterial meningitis; <strong>of</strong>ten the bacteria can be directly characterized.<br />

Prevalence <strong>of</strong> lymphatic cells with moderate pleocytosis suggests viral<br />

meningitis. (If clinical and serological findings leave doubts, the differential<br />

diagnosis must rule out lymphoma using immunocytological<br />

methods.)<br />

Strong eosinophilia suggests parasite infection (e.g., cysticercosis).<br />

A complete mixture <strong>of</strong> cells with granulocytes, lymphocytes, and monocytes<br />

in equal proportion is found in tuberculous meningitis.<br />

Variable blasts, usually with significant pleocytosis, predominate in<br />

leukemic or lymphomatous meningitis.<br />

Undefinable cells with large nuclei suggest tumor cells in general, e.g.,<br />

meningeal involvement in breast cancer or bronchial carcinoma, etc. The<br />

cell types are determined on the basis <strong>of</strong> knowledge <strong>of</strong> the primary tumor<br />

and/or by marker analysis. Among primary brain tumors, the most likely<br />

cells to be found in cerebrospinal fluid are those from ependymoma,<br />

pinealoma, and medulloblastoma.<br />

Erythrophages and siderophages (siderophores) are monocytes/macrophages,<br />

which take up erythrocytes and iron-containing pigment during<br />

subarachnoid hemorrhage.<br />

The cytological analysis <strong>of</strong> the cerebrospinal fluid <strong>of</strong>fers important clues<br />

to the character <strong>of</strong> meningeal inflammation, the presence <strong>of</strong> a malignancy,<br />

or hemorrhage.<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


a<br />

c<br />

e<br />

Viral, bacterial, and malignant meningitis can be distinguished<br />

by means <strong>of</strong> cerebrospinal fluid cytology<br />

g<br />

h<br />

Fig. 68 Cerebrospinal fluid cytology. a Cerebrospinal fluid cytology in bacterial<br />

meningitis: granulocytes with phagocytosed diplococci (in this case, pneumococci,<br />

arrow). b Cerebrospinal fluid cytology in viral meningitis: variable lymphoid<br />

cells. c Cerebrospinal fluid cytology in non-Hodgkin lymphoma: here, mantle cell<br />

lymphoma. d Cerebrospinal fluid cytology after subarachnoid hemorrhage: macrophages<br />

with phagocytosed erythrocytes. e–h Cerebrospinal fluid cytology in<br />

meningeal involvement in malignancy: the origin <strong>of</strong> the cells cannot be deduced<br />

with certainty from the spinal fluid cytology alone: (e) breast cancer, (f) bronchial<br />

carcinoma, (g) medulloblastoma, and (h) acute leukemia.<br />

189<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.<br />

b<br />

d<br />

f


190<br />

References<br />

Begemann, H., M. Begemann: Praktische Hämatologie, 10. Aufl. Thieme, Stuttgart<br />

1998<br />

Begemann, H., J. Rastetter: Klinische Hämatologie, 4. Aufl. Thieme, Stuttgart 1993<br />

Bessis, M.: Blood Smears Reinterpreted. Springer, Berlin 1977<br />

Binet, J.L., A. Auquier, G. Dighiero et al.: A new prognostic classification <strong>of</strong> chronic<br />

lymphocytic leukemia derived from a multivariate survival analysis. Cancer<br />

1981; 48(1): 198-206<br />

Brücher, H.: Knochenmarkzytologie. Thieme, Stuttgart 1986<br />

Classen, M., A. Dierkesmann, H. Heimpel et al.: Rationelle Diagnostik und Therapie<br />

in der inneren Medizin. Urben & Schwarzenberg, München 1996<br />

Costabel, M.: <strong>Atlas</strong> der bronchoalveolären Lavage. Thieme, Stuttgart 1994<br />

Costabel V., J. Guzman: Bronchoalveolar lavage in interstital lung disease. Curr<br />

Opin Pulm Med 2001; 7(5): 255-61<br />

Durie, B.G.M., S.E. Salmon: A clinical staging system for multiple myeloma. Correlation<br />

<strong>of</strong> measured myeloma cell mass with presenting clinical features, response<br />

to treatment, and survival. Cancer 1975; 36: 842-854<br />

Heckner, F., M. Freund: Praktikum der mikroskopischen Hämatologie. Urban &<br />

Schwarzenberg, München 1994<br />

Heimpel, H., D. Hoelzer, E. Kleihauer, H. P. Lohrmann: Hämatologie in der Praxis.<br />

Fischer, Stuttgart 1996<br />

Huber, H., H. Löffler, V. Faber: Methoden der diagnostischen Hämatologie.<br />

Springer, Berlin 1994<br />

Jaffe, E. S., N. L. Harris, H. Stein, J. W. Vardiman: World Health Organization Classification<br />

<strong>of</strong> Tumours. Pathology and Genetics <strong>of</strong> Tumours <strong>of</strong> Haematopoietic and<br />

Lymphoid Tissues. IARC Press, Lyon 2001<br />

Lennert, K., A. C. Feller: Non-Hodgkin-Lymphome. Springer, Berlin 1990<br />

Löffler, H., J. Rastetter: <strong>Atlas</strong> der klinischen Hämatologie. Springer, Berlin 1999<br />

Murphy, S.: Diagnostic criteria and prognosis in polycythemia vera and essential<br />

thombocythemia. Semin Hematol 1999; 36(1 Suppl 2): 9-13<br />

Ovell, S. R., G. Sterrett, M. N. Walters, D. Whitaker: Fine Needle Aspiration Cytology.<br />

Churchill Livingstone, Edinburgh London 1992<br />

Pearson, T.C., M Messinezy: The diagnostic criteria <strong>of</strong> polycythaemia rubra vera.<br />

Leuk Lymphoma 1996;22(Suppl 1): 87-93<br />

Pralle, H. B.: Checkliste Hämatologie, 2. Aufl. Thieme, Stuttgart 1991<br />

Schmoll, H. J., K. Höffken, K. Possinger: Kompendium internistische Onkologie,<br />

3. Bde., 3. Aufl. Springer, Berlin 1999<br />

Theml, H., W. Kaboth, H. Begemann: Blutkrankheiten. In Kühn, H. A., J. Schirmeister:<br />

Innere Medizin, 5. Aufl. Springer, Berlin 1989<br />

Theml, H., H. D. Schick: Praktische Differentialdiagnostik hämatologischer und<br />

onkologischer Krankheiten. Thieme, Stuttgart 1998<br />

Zucker-Franklin, D., M. F. Greaves, C. E. Grossi, A. M. Marmont: <strong>Atlas</strong> der Blutzellen,<br />

2. Aufl. Fischer, Stuttgart 1990<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Index<br />

A<br />

Actinomycosis 179<br />

Addison disease 66<br />

Agammaglobulinemia 48<br />

Agranulocytosis 48, 86<br />

acute 86<br />

allergic 8<br />

bone marrow analysis 54, 86, 87<br />

classification 86<br />

subacute 86<br />

Alcoholism, chronic 168<br />

Allergic processes 8, 44<br />

basophilia and 124<br />

eosinophilia and 124<br />

Anemia 80, 106–107<br />

aplastic 8, 48, 56, 131, 140, 148–150<br />

bone marrow analysis 53, 54, 56<br />

classification 128<br />

congenital dyserythropoietic (CDA)<br />

147, 148<br />

diagnosis 130–131<br />

Diamond–Blackfan 146<br />

dimorphic 152<br />

hemolytic 131, 140–145<br />

causes 142<br />

microangiopathic 144, 150<br />

with erythrocyte anomalies<br />

144–145<br />

hemorrhage and 131<br />

hyperchromic 128, 152–155<br />

causes 152<br />

hyper-regenerative 128<br />

hypochromic 128–139<br />

bone marrow analysis 134–137<br />

infectious/toxic 131, 134–135, 136<br />

iron-deficiency 128–133, 136<br />

sideroachrestic 137<br />

with hemolysis 138–139<br />

hypoplastic 140<br />

congenital 146<br />

hyporegenerative 128<br />

immunohemolytic 8<br />

macrocytic 152–153<br />

191<br />

megaloblastic 43, 54, 56, 131,<br />

152–154<br />

normochromic 128, 140–151<br />

bone marrow analysis 140<br />

hemolytic 140–145<br />

renal 146<br />

paraneoplastic 131<br />

pernicious 140, 154<br />

refractory 106<br />

in transformation 108<br />

with excess blasts (RAEB) 54, 106<br />

with ring sideroblasts 106<br />

renal 146<br />

secondary 131, 134, 135, 136<br />

sickle cell 142, 144, 145<br />

sideroachrestic 131, 137<br />

sideroblastic 137<br />

smoldering 54<br />

Anisocytosis 130, 134–135, 137–139,<br />

152–153, 170<br />

Anulocytes 132, 133<br />

Ascites 186–187<br />

Auer bodies 89, 96–99<br />

Autoantibodies 142<br />

Autoimmune processes 8, 180<br />

anemia and 134<br />

autoagglutination 142, 143<br />

eosinophilia and 124<br />

Automated blood count 19–20<br />

Azurophilic granules 96, 100<br />

B<br />

B-lymphocytes 48, 49<br />

function 6<br />

see also Lymphocytes<br />

Band cells 4, 6, 38–39, 57, 153<br />

diagnostic implications 38, 112, 113<br />

normal ranges and mean values 12<br />

Basophilia 124–126<br />

Basophilic stippling 134, 135,<br />

137–139, 156–157<br />

Basophils 44–45, 125<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


192<br />

Index<br />

Basophils, diagnostic implications 44<br />

elevated counts 124–126<br />

function 5, 6<br />

in chronic myeloid leukemia 118,<br />

120, 121<br />

normal ranges and mean values 13<br />

Bicytopenia 106, 149<br />

Blast crisis<br />

in chronic myeloid leukemia<br />

120–121<br />

in osteomyelosclerosis 123<br />

Blood cell series 2–5, 53<br />

regulation and dysregulation 7–8<br />

see also Erythropoiesis; Granulocytopoiesis<br />

Blood counts<br />

automated blood counts 19–20<br />

complete blood count (CBC) 25<br />

differential diagnosis 62–65<br />

differential blood count (DBC)<br />

17–19, 25–26<br />

indications 27<br />

erythrocytes 10, 13, 25<br />

leukocytes 14, 16, 25<br />

reticulocytes 11, 13, 32<br />

thrombocytes 15, 25<br />

Blood sample collection 9<br />

Blood smear 17–19<br />

staining 18, 19<br />

Blood–bone marrow barrier, destruction<br />

<strong>of</strong> 30, 32, 34<br />

Boeck disease 178, 180, 181<br />

Bone marrow 52–59<br />

analysis 52–59<br />

agranulocytosis 54, 86, 87<br />

cell density 52<br />

chromic myeloid leukemia<br />

118–119, 120, 121<br />

essential thrombocythemia<br />

170–171<br />

hypochromic anemias 134–137<br />

indications for 26, 27<br />

myelodysplasias 108, 109<br />

normochromic anemias 140<br />

polycythemia vera 162–163<br />

red cell series/white cell series<br />

ratios 53, 136<br />

space-occupying processes<br />

150–151<br />

thrombocytopenia 164, 169<br />

aplasia 146–150<br />

pure red cell aplasia (PRCA)<br />

146–148<br />

biopsy 20–22, 27<br />

cytology 20, 52–55<br />

disturbances 8<br />

histology 20, 26<br />

hyperplasia 149<br />

infectious/toxic 134<br />

medullary stroma cells 58–59<br />

neoplasms 150, 168<br />

carcinosis 131, 150–151<br />

Bone metastases 150, 168<br />

Branchial cyst 179, 184, 185<br />

Bronchoalveolar lavage 184–185<br />

indications 184<br />

Brucellosis 66, 179<br />

Bruton disease 48<br />

Burkitt lymphoma 71<br />

Burr cell 135<br />

C<br />

Cabot ring 156, 157<br />

Carcinoma 188, 189<br />

bone marrow 131, 150–151<br />

metastatic disease 182, 183<br />

Cat-scratch disease 179<br />

Centroblasts 176<br />

Cerebrospinal fluid 188–189<br />

Charcot-Leyden crystals 44<br />

Chickenpox 66<br />

Chromatin structure 4<br />

Collagen disease 179<br />

Complete blood count (CBC) 25<br />

differential diagnosis 62–65<br />

Cyclic Murchison syndrome 40<br />

Cyst<br />

branchial 179, 184, 185<br />

retention 184<br />

Cytomegalovirus (CMV) infection 66,<br />

67, 68, 168<br />

D<br />

Diagnostic workup 25–27<br />

Diamond–Blackfan anemia 146<br />

Differential blood count (DBC) 17–19,<br />

25–26<br />

indications 27<br />

DiGeorge disease 48<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


Disseminated intravascular<br />

coagulopathy (DIC) 144, 167<br />

Döhle bodies 39–41<br />

Drumstick appendages 42, 43<br />

Ductus thyroglossus 184<br />

Dyserythropoiesis 102, 106, 109<br />

congenital dyserythropoietic anemia<br />

(CDA) 147, 148<br />

Dysgranulopoiesis 102, 106<br />

Dysmegakaryopoiesis 102, 106, 109<br />

E<br />

Elliptocytosis 142<br />

Eosinophilia 111, 124, 125<br />

cerebrospinal fluid 188<br />

Eosinophils 44–45, 125<br />

diagnostic implications 44, 56<br />

elevated counts 124, 125<br />

functions 5, 6<br />

in chronic myeloid leukemia 118<br />

normal ranges and mean values 12<br />

Epstein–Barr virus (EBV) infection 66,<br />

68–69, 168<br />

Erythremia 54, 162–163<br />

Erythroblastopenia 146–148<br />

Erythroblastophthisis 146<br />

Erythroblastosis 8<br />

Erythroblasts 30–33, 57<br />

acute erythroleukemia 100<br />

basophilic 30, 32, 54, 57, 85<br />

diagnostic implications 30, 32<br />

orthochromatic 31, 32, 54, 57<br />

polychromatic 32–33, 35, 54, 57<br />

Erythrocytes 7, 33<br />

assessment 26<br />

basophilic stippling 134, 135,<br />

137–139, 156–157<br />

counts 10, 13, 25, 128<br />

function 7<br />

inclusions 156–161<br />

iron deficiency and 132–133<br />

normal ranges and mean values 13<br />

parameters 10–11<br />

red cell distribution width (RDW)<br />

11<br />

ring-shaped 130, 132, 133<br />

tear-drop 115, 123<br />

see also Anemia<br />

Erythrocytosis<br />

primary 162<br />

193<br />

secondary 162, 163<br />

Erythroleukemia 30, 93, 100<br />

in normochromic anemia 140<br />

Erythrophages 188<br />

Erythropoiesis 3, 30–33, 55<br />

bone marrow analysis 54, 136<br />

iron staining 56, 57, 109<br />

dyserythropoiesis 102, 106, 109<br />

in hypochromic anemia 136, 138<br />

in normochromic anemia 140<br />

megaloblastic 54<br />

Erythropoietin 146<br />

Essential thrombocythemia (ET) 8, 26,<br />

56, 114, 170–171<br />

diagnosis 170<br />

F<br />

Faggot cells 99<br />

Felty syndrome 179<br />

Ferritin granules 56<br />

Fibroblastic reticular cells 58, 59<br />

Folic acid deficiency 7, 38, 54, 149<br />

in hemolytic anemia 140, 141<br />

in hyperchromic anemia 152–154<br />

thrombocytopenia and 168<br />

Fragmentocytes 142, 143<br />

Fragmentocytosis 144<br />

G<br />

Index<br />

Gaisböck syndrome 53<br />

Gaucher syndrome 118<br />

Granulocytes 3, 4, 12, 13<br />

degrading forms 42<br />

functions 5, 6<br />

hypersegmented 153–154<br />

precursors 34–37<br />

see also Basophils; Eosinophils;<br />

Neutrophils<br />

Granulocytopenia 66, 86<br />

Granulocytopoiesis 3, 5, 34–39<br />

bone marrow analysis 54<br />

dysgranulopoiesis 102, 106<br />

in acute myelomonocytic leukemia<br />

98<br />

in hypochromic anemia 136<br />

Granulocytosis 110<br />

Gumprecht's nuclear shadow 74<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


194<br />

H<br />

Heinz bodies 142, 156<br />

HELLP syndrome 144<br />

Hematocrit 10<br />

Hematopoiesis<br />

extramedullary 30, 32<br />

see also Erythropoiesis<br />

Hemoglobin<br />

assay 10, 128<br />

deficiency 132<br />

mean cell hemoglobin content<br />

(MCH) 10, 13, 128<br />

mean cellular hemoglobin concentration<br />

(MCHC) 10, 11, 13<br />

normal ranges and mean values 13<br />

see also Anemia<br />

Hemoglobinopathies 144<br />

Hemolysis 32, 54, 138–139, 142–143<br />

Hemolytic uremic syndrome (HUS)<br />

144<br />

Hemophagocytosis 46<br />

HEMPAS antigen 148<br />

Hepatitis 66<br />

Histiocytes 118<br />

reticular 58<br />

sea-blue 118<br />

Hodgkin cells 176, 177<br />

Hodgkin disease 26, 70, 176, 177<br />

eosinophilia 124<br />

Howell–Jolly bodies 139, 156, 157<br />

Hypereosinophilia syndrome 124<br />

Hypergammaglobulinemia 82, 131<br />

differential diagnosis 82<br />

Hypersensitivity reactions 44<br />

eosinophilia and 124<br />

Hyperthyroidism 66<br />

Hypogammaglobulinemia 74, 82<br />

I<br />

Index<br />

IgM paraprotein 78<br />

Immunoblasts 176<br />

Immunocytoma, lymphoplasmacytoid<br />

70–71, 74, 77–78, 183<br />

Immunothrombocytopenia 165<br />

drug-induced 164–166<br />

idiopathic 166<br />

secondary 166<br />

Infection 36, 40, 46, 48, 112–113<br />

anemia and 134<br />

basophilia and 124<br />

eosinophilia and 124<br />

thrombocytopenia and 168<br />

see also specific infections<br />

Infectious lymphocytosis 66<br />

Infectious mononucleosis 68–69<br />

Influenza 168<br />

Iron<br />

deficiency 7, 56, 128–133<br />

blood cell analysis 132<br />

bone marrow analysis 136<br />

normal ranges 132<br />

Iron staining 56, 57, 109<br />

Isoantibodies 142<br />

L<br />

Langhans giant cells 180, 181<br />

Leukemia 8, 34, 40, 90–91<br />

acute 90–105<br />

basophilic 126<br />

bone marrow analysis 54<br />

characteristics 96<br />

classifications 91–93, 94, 104<br />

diagnosis 91–94<br />

eosinophilic 124<br />

erythroleukemia 30, 93, 100<br />

lymphocytic (ALL) 104–105<br />

classification 104<br />

mast cell 126<br />

megakaryoblastic 102<br />

megakaryocytic 93<br />

monoblastic 93, 100–101<br />

monocytic 46, 89, 93, 100<br />

myeloblastic 96<br />

myeloid (AML) 89, 92, 94–97<br />

hypoplastic 102, 103<br />

with dysplasia 102, 103<br />

myelomonocytic 92, 98, 99<br />

promyelocytic 92, 98, 99<br />

aleukemic 149<br />

B-prolymphocytic (B-PLL) 70, 74, 77<br />

chronic<br />

basophilic 126<br />

lymphocytic (CLL) 66, 70, 74–79,<br />

90<br />

characteristics 76<br />

staging 76, 78<br />

myeloid (CML) 26, 44, 56, 90,<br />

114–121<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


last crisis 120–121<br />

bone marrow analysis 118–119,<br />

120, 121<br />

characteristics 116, 124<br />

diagnosis 54, 111, 116–119<br />

myelomonocytic (CMML) 89, 90,<br />

107, 108<br />

eosinophilic 124<br />

erythroleukemia 30<br />

hairy cell (HCL) 71, 80, 81<br />

variant (HCL-V) 80<br />

lymphoplasmacytic 71<br />

megalokaryocytes 50<br />

peroxidase-negative 91<br />

peroxidase-positive 91<br />

plasma cell 48, 78, 82<br />

pseudo-Pelger granulocytes 42<br />

T-prolymphocytic 74, 77<br />

Leukemic hiatus 91<br />

Leukocyte alkaline phosphatase 54<br />

Leukocytes 3–4<br />

counts 14, 16, 25, 90–91<br />

normal ranges and mean values 12<br />

Leukocytopenia 80, 107<br />

Leukocytosis 63, 110<br />

smoker's 111<br />

Leukopenia 63<br />

Listeriosis 179<br />

Loeffler syndrome 124<br />

Lung aspirates 186–187<br />

Lymph node biopsy 23, 24, 174–183<br />

metastatic disease 182, 183<br />

non-Hodgkin lymphoma 182, 183<br />

reactive lymph node hyperplasia<br />

176–179<br />

sarcoidosis 180–181<br />

tuberculosis 180–181<br />

Lymphadenitis<br />

reactive 176, 178–179<br />

tuberculous 181<br />

Lymphadenoma 54, 149<br />

chronic 74<br />

marginal zone 78, 80<br />

Lymphatic cells 63, 66<br />

cerebrospinal fluid 188<br />

differentiation in non-Hodgkin<br />

lymphoma 72–73<br />

reactive lymphocytosis 66–67<br />

see also Lymphocytes<br />

Lymphoblasts 32, 176<br />

acute lymphoblastic leukemia (ALL)<br />

104–105<br />

195<br />

Lymphocytes 3, 4, 33, 48–49<br />

bone marrow 56<br />

diagnostic implications 48, 56, 66<br />

functions 6<br />

in infectious mononucleosis 68–69<br />

large granulated (LGL) 69<br />

normal ranges and mean values 12<br />

plasmacytoid 77, 176<br />

see also Lymphocytosis; Lymphoma<br />

Lymphocytopoiesis 3<br />

Lymphocytosis 66–69, 74, 76<br />

constitutional relative 66<br />

infectious 66<br />

reactive 66–67, 86–87<br />

Lymphogranulomatosis see Hodgkin<br />

disease<br />

Lymphoma 69, 77, 82, 174<br />

B-cell 70, 71<br />

Burkitt 71<br />

centroblastic 71<br />

centrocytic 71<br />

classification 70–72<br />

follicular 71, 78, 79<br />

large-cell 71, 74<br />

lymphoblastic 72<br />

lymphoplasmacytic 78<br />

malignant 26<br />

mantle cell 71, 78, 79, 189<br />

marginal zone 71<br />

monocytoid 71<br />

non-Hodgkin (NHL) 26, 70–73<br />

blastic 70<br />

cell surface markers 72–73<br />

lymph node biopsy 182, 183<br />

precursor 70<br />

reactive 176–179<br />

diagnosis 178–179<br />

splenic lymphoma with villous<br />

lymphocytes (SLVL) 80, 81<br />

T-cell 72<br />

cutaneous (CTCL) 74, 77<br />

Lymphomatous toxoplasmosis 66<br />

M<br />

Index<br />

Macroblasts 30<br />

Macrocytes 130, 153<br />

Macrophages 5–6, 59<br />

iron-storage 57, 58, 136<br />

role in erythropoiesis 30, 32<br />

Malaria 19, 158–161<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


196<br />

Index<br />

Malignant lymphogranulomatosis 70<br />

Malignant transformations 8<br />

Mastocytosis 125, 126<br />

May-Hegglin anomaly 41, 166, 168<br />

Mean cell hemoglobin content (MCH)<br />

10, 13, 128<br />

Mean cell volume (MCV) 10, 13<br />

Mean cellular hemoglobin concentration<br />

(MCHC) 10, 11, 13<br />

Measles 66, 166, 168<br />

Medulloblastoma 188, 189<br />

Megakaryocytes 50–51, 57<br />

diagnostic implications 50, 56<br />

in chronic myeloid leukemia 115,<br />

118<br />

in essential thrombocythemia<br />

170–171<br />

in hyperchromic anemia 154<br />

in iron deficiency 136<br />

in osteomyelosclerosis 122<br />

Megaloblasts 154<br />

Megalocytes 130, 152, 153<br />

Meningitis 188, 189<br />

Merozoites 158<br />

Metabolite deficiencies 7<br />

Metamyelocytes 4, 31, 35–37, 57, 153<br />

diagnostic implications 36<br />

Metastases<br />

bone 150, 168<br />

lymph nodes 182, 183<br />

Microangiopathy 144<br />

thrombocytopenia in 166<br />

Microcytes 132, 133<br />

Micromegakaryocytes 118<br />

Micromyeloblasts 34<br />

Microspherocytosis 144, 145<br />

Monocytes 3, 41, 46–47<br />

acute monocytic leukemia 100<br />

diagnostic implications 46, 56<br />

functions 5–6<br />

normal ranges and mean values 12<br />

Monocytopoiesis 3<br />

in acute myelomonocytic leukemia<br />

98<br />

Monocytosis 46, 88–89<br />

causes 88<br />

Mononucleosis 66, 68–69, 168, 176,<br />

178<br />

Multiple myeloma (MM) 82, 83, 85<br />

Mumps 166<br />

Myeloblasts 4, 31, 34–35<br />

acute erythroleukemia 100<br />

acute myeloblastic leukemia 96–97<br />

diagnostic implications 34<br />

see also Blast crisis<br />

Myelocytes 4, 36–37, 57<br />

diagnostic implications 36<br />

in hyperchromic anemia 154<br />

Myelodysplasia (MDS) 40, 42,<br />

106–109, 131, 149<br />

bone marrow analysis 54, 108, 109<br />

classification 106, 108<br />

differential diagnosis 154–155<br />

Myel<strong>of</strong>ibrosis (MF) 26<br />

Myeloma<br />

multiple (MM) 82, 83, 85<br />

plasma cell 71, 82, 84<br />

Myeloproliferative diseases 32, 44, 50,<br />

56, 149<br />

basophilia and 124–126<br />

chronic myeloproliferative disorders<br />

(CMPD) 114–115<br />

differential diagnosis 115<br />

see also specific diseases<br />

N<br />

Natural killer (NK) cells 48<br />

Neutropenia 86<br />

autoimmune 86<br />

classification 86<br />

congenital/familial 86<br />

secondary, in bone marrow disease<br />

86<br />

Neutrophilia 110<br />

causes 111, 112<br />

reactive left shift 112–113<br />

without left shift 110<br />

Neutrophils 12, 31, 38–41, 45<br />

cerebrospinal fluid 188<br />

functions 5, 6<br />

segmented 38–41, 43<br />

diagnostic implications 38<br />

normal ranges and mean values<br />

12<br />

see also Neutropenia; Neutrophilia<br />

Non-Hodgkin lymphoma (NHL) 26,<br />

70–73<br />

blastic 70<br />

cell surface markers 72–73<br />

lymph node biopsy 182, 183<br />

Normal values 15–17<br />

Normoblasts 32, 138, 150<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


in hemolytic anemia 140<br />

orthochromatic 140<br />

Nuclear appendages 42, 43<br />

O<br />

Osteoblasts 58, 59<br />

Osteoclasts 58, 59<br />

Osteomyelosclerosis (OMS) 26, 111,<br />

114, 122–123<br />

characteristics 122<br />

Oversegmentation 38<br />

P<br />

Pancytopenia 80<br />

Panmyelopathy 56, 148–149<br />

causes 149<br />

Panmyelophthisis 8, 148–150, 168<br />

Parasite infections 44, 124, 188<br />

eosinophilia and 124, 188<br />

Pel–Ebstein fever 40<br />

Pelger anomaly 40<br />

Perls' Prussion blue reaction 56<br />

Pertussis 66<br />

Pfeiffer cells 68, 69<br />

Philadelphia chromosome 114, 115,<br />

116<br />

Plasma cells 48–49, 57, 77, 82–85<br />

counts 56<br />

diagnostic implications 48, 82<br />

Plasmablasts 176<br />

Plasmacytoma 56, 70, 71, 82–85, 149<br />

morphological variability 84<br />

staging 84<br />

Plasmodia 19, 158–161<br />

Pleural effusions 186–187<br />

Poikilocytosis 130, 134, 135, 137–139,<br />

152<br />

Polychromasia 135<br />

Polychromophilia 134, 137<br />

Polycythemia 8, 53, 56, 111, 131<br />

erythremic 162–163<br />

Polycythemia vera (PV) 114, 122,<br />

162–163<br />

diagnosis 162<br />

rubra 26<br />

Proerythroblasts 30–31, 54, 85<br />

Promyelocytes 4, 31, 34–35, 44, 57<br />

acute promyelocyte leukemia 98, 99<br />

197<br />

diagnostic implications 34, 83<br />

Pseudo Gaucher cells 118<br />

Pseudo-Pelger formation 40, 41, 107<br />

diagnostic implications 42, 118<br />

Pseudopolycythemia 53<br />

Pseudothrombocytopenia 15, 51, 164,<br />

166–167<br />

Q<br />

5q-syndrome 108<br />

R<br />

Rabbit fever 179<br />

Reactive lymph node hyperplasia 176,<br />

177<br />

Reactive lymphocytosis 66–67<br />

Red cell distribution width (RDW) 11<br />

Reed–Sternberg giant cells 176, 177<br />

Reference ranges 15–17<br />

Refractive anemia with excess blasts<br />

(RAEB) 54<br />

Renal insufficiency 146<br />

Reticular cells, fibroblastic 58, 59<br />

Reticulocytes 32<br />

counts 11, 13, 32, 128<br />

in hemolytic anemia 140–141<br />

normal ranges and mean values 13<br />

Reticuloendothelial system (RES), iron<br />

pull 134, 136<br />

Richter syndrome 74, 77<br />

Rubella 66, 166, 168, 178<br />

Russell bodies 84, 85<br />

S<br />

Sarcoidosis 178, 180–181<br />

Scarlet fever 40<br />

Schistocytosis 144<br />

Schizocytes 142<br />

Schizonts 158<br />

Schüffner's dots 158<br />

Scr<strong>of</strong>uloderma 180<br />

Sepsis 40, 41, 113, 167<br />

Sézary syndrome 74, 77<br />

Sickle cells 142, 144, 145<br />

Sideroachresia 56, 137<br />

Sideroblasts 56, 137<br />

Index<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.


198<br />

Sideroblasts, ring 56, 106, 109, 137<br />

Siderophages 188<br />

Spherocytosis 142<br />

Splenic lymphoma with villous<br />

lymphocytes (SLVL) 80, 81<br />

Splenomegaly 80, 112, 114–116, 142<br />

in chronic myeloid leukemia<br />

114–115, 116<br />

in osteomyelosclerosis 123<br />

Staff cells 4<br />

Staining 18, 19<br />

iron staining <strong>of</strong> erythropoietic cells<br />

56, 57, 109<br />

Stem cells 2–3<br />

Still disease 179<br />

Stomatocytes 144, 145<br />

Stomatocytosis 144<br />

Strongyloides stercoralis 124<br />

Subarachnoid hemorrhage 188, 189<br />

Substantia granul<strong>of</strong>ilamentosa 156<br />

T<br />

Index<br />

T-lymphocytes 48<br />

function 6<br />

see also Lymphocytes<br />

Target cells 130, 138, 139, 142<br />

Thalassemia 131, 138–139, 142<br />

major 138, 139<br />

minor 138, 139<br />

Thrombocytes 7, 33, 50–51, 165–169<br />

counts 15, 25<br />

diagnostic implications 50<br />

function 7<br />

normal ranges and mean values 13<br />

Thrombocythemia<br />

essential (ET) 8, 26, 56, 114,<br />

170–171<br />

diagnosis 170<br />

idiopathic 122<br />

Thrombocytopenia 8, 56, 80, 113,<br />

164–169<br />

due to increased demand 164–167<br />

due to reduced cell production<br />

168–169<br />

in chronic myeloid leukemia 121<br />

in hypersplenism 166<br />

in microangiopathy 166<br />

in myelodysplasia 107<br />

Thrombocytopenic purpura (TTP) 131,<br />

144, 166<br />

post-transfusion 166<br />

Thrombocytosis 170–171<br />

reactive 170<br />

Thrombopoiesis 3<br />

Tissue mast cells 125, 126<br />

Toxic effects 8<br />

anemia 134<br />

Toxic granulation 40–41, 113<br />

diagnostic implications 42<br />

Toxoplasmosis 176, 178, 180<br />

lymphomatous 66<br />

Tricytopenia 102, 106, 149<br />

Trophozoites 158, 161<br />

Tuberculosis 178, 180–181<br />

Tularemia 179<br />

Tumors<br />

anemia and 134<br />

biopsy 23<br />

see also specific tumors<br />

U<br />

Urticaria pigmentosa 126<br />

V<br />

Vacuoles 40, 41<br />

Virocytes 66, 68, 69<br />

Vitamin B12 deficiency 7, 38, 54, 149<br />

in hyperchromic anemia 152–154<br />

thrombocytopenia and 168<br />

W<br />

Waldenström syndrome 78<br />

Werlh<strong>of</strong> syndrome 56<br />

Wheel-spoke nuclei 84<br />

Whooping cough 66<br />

Wiscott-Aldrich syndrome 168<br />

X<br />

X-chromosome 42, 43<br />

Z<br />

Zieve syndrome 142<br />

Theml, <strong>Color</strong> <strong>Atlas</strong> <strong>of</strong> <strong>Hematology</strong> © <strong>2004</strong> Thieme<br />

All rights reserved. Usage subject to terms and conditions <strong>of</strong> license.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!